CN106061982A - Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof - Google Patents

Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof Download PDF

Info

Publication number
CN106061982A
CN106061982A CN201480074587.9A CN201480074587A CN106061982A CN 106061982 A CN106061982 A CN 106061982A CN 201480074587 A CN201480074587 A CN 201480074587A CN 106061982 A CN106061982 A CN 106061982A
Authority
CN
China
Prior art keywords
compound
experimenter
virus
pharmaceutically acceptable
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201480074587.9A
Other languages
Chinese (zh)
Inventor
罗伊·韦尔
亚伦·唐尼
欧内斯特·拉尼尔
菲罗泽·塞斯纳
迪恩·塞列斯泽
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chimerix Corp
Original Assignee
Chimerix Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chimerix Corp filed Critical Chimerix Corp
Publication of CN106061982A publication Critical patent/CN106061982A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/6512Six-membered rings having the nitrogen atoms in positions 1 and 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present invention is directed to branched chain nucleoside phosphonate ester compounds and methods of synthesis thereof. The present invention is also directed to pharmaceutical compositions comprising branched chain nucleoside phosphonate ester compounds and methods of treating and/or preventing double stranded DNA viral infection and/or viral infection associated disease or disorder.

Description

Branched-chain acyclic nucleoside phosphate ester and synthetic method thereof and application
Related application
This application claims and enjoy the preferential of No. 61/912,407 U.S. Provisional Patent Application that December in 2013 submits on the 5th Power and interests, during entire contents is incorporated herein by reference by entirety.
Technical field
The application relates to branched-chain acyclic nucleoside phosphonate compound, its analog, its pharmaceutical composition and synthetic method thereof. The invention still further relates to infect by the described sweet phosphonate compound of branched-chain acyclic core, its derivant and medicine composite for curing virus thereof Method.
Background technology
Virus infects and individual and entire society can be produced harmful effect.Except deadly virus infects such as Ebola, even Non-lethal infection is likely to produce serious economic consequences.Such as, 1999, at the direct medical care expenses of the U.S.'s only influenza infection With just account for 10-30 hundred million dollars, not to mention the indirect expense of 100-150 hundred million dollars.See Szucs, J.Antimicrob.Chemother. (1999), Topic B, 11-15.The such as human cytomegalovirus of virus additionally (HCMV), BK virus (BKV), epstein-Barr virus (EBV), adenovirus, JC virus (JCV), SV40, MC virus (MCV), KI virus (KIV), WU virus (WUV), cowpox, herpes simplex virus type 1 (HSV-1), herpes simplex virus type 2 (HSV-2), human herpes virus-6 (HHV-6), Human herpesvirus 8 (HHV-8), hepatitis B virus, hepatitis C virus Poison, varicella zoster virus (VZV), classical smallpox, alastrim, variola, cowpox, camel pox, monkeypox, poliomyelitis disease Poison, Ebola virus, Marburg virus, enterovirus, papillomavirus and HIV (human immunodeficiency virus) (HIV) can be each From producing significantly society and economic impact.
Therefore, the development of effective antiviral therapy of these viruses it is effective against to improving infected individuals such as transplant patient Health status is important, and prevents the outbreak of other pathogenic virus as a kind of public health measure.
Nucleoside phosphate ester (such as, ribonucleotide derivative) represents and depends on use ribonucleotide is that substrate virus is compiled The target class antiviral drugs of the mould a kind of excellence suppressing virus of code, such as some varial polymerases to many RNA viruses And/or to RNA or the viral helicase of DNA viruses.But, an obstacle of this kind of antiviral drugs curative effect is that needs are thin at target The bio-modification of intracellular oral formulations, to form reactive antiviral medicine.If a nucleoside is carried, need three phosphorylations Step forms triphosphate.First phosphorylation is walked around in the conveying of nucleoside phosphate ester effectively, but exacerbates through lipid The problem that cell delivery around bilayer has the charged drugs of consumption clinically.
Lipid binding can be used to pretend oral drugs (including nucleoside phosphate ester) and is easily absorbed by the body as a kind of Native compound.Specifically, nucleoside phosphate ester can be modified into the phospholipid of similar portions metabolism (monoacyl).With normally Diacyl phosphatidyl is contrary, and the nucleoside of monoacyl lipid modification can readily penetrate through the inner chamber of intestinal lining intestinal, enters blood circulation And/or lymph, unlike standard drug, keep complete.Therefore, lipid part the most not only provides nucleoside to blood plasma;It promotes Efficient absorption is to target cell.Lipid is to crack at the Cytoplasm compartment of target cell and in the case of nucleoside analog conjugates, Obtain corresponding monophosphate.In general, this strategy may result in the reactive antiviral medicine water at the position of virus replication Flat is greatly increased.
Present invention accomplishes by using new antiviral drugs and delivery vehicles can be used for treatment and/or prevent to cause The demand of the new therapy of the virus of disease.
Part of the present invention provides the branched nucleosides phosphonate ester as antiviral drugs and synthetic method thereof.The present invention is open Additionally provide use one or more embodiments compound treat and/or prophylaxis of viral infections with or relevant with virus infection Disease or the method for disease.
Summary of the invention
The present invention relates to the compound of formula (I)
With its pharmaceutically acceptable enantiomer, diastereomer, racemic modification, mixture or salt, wherein R is:
The invention still further relates to following compounds:
With its pharmaceutically acceptable salt.
One embodiment of the present of invention relates to the typical compound of formula (I) Synthetic method, shown in following option A.
Option A:
Such as, described method comprises the steps:
I () adds magnesium chips in 2-methyltetrahydrofuran (Me-THF) solution of 1-bromo-3-methybutane;
(ii) in reactant mixture, add the Me-THF solution of the bromo-DODECANOL, 1-of 12-, be subsequently added four chloro copper acid Oxolane (THF) solution of two lithiums, to synthesize compound 3;
(iii) in compound 3 and N, the cold soln of the dichloromethane of N-diisopropylethylamine (DIPEA), methylsulfonyl is added Chlorine, keeps temperature to be below about 5 DEG C, to provide compound 4 simultaneously;
(iv) in the cold soln of the METHYLPYRROLIDONE (NMP) of 1,3-PD, sodium hydride (NaH) is added, with Rear addition compound 4, to synthesize compound 5;
V () adds bromotrimethylsilane (TMS-Br) in the acetonitrile solution of compound 6 (purchased from Lacamas laboratory);
(vi), after removing acetonitrile and TMS-Br, ethanedioly chloride and dimethylformamide (DMF) are added, to form compound 7;
(vii) in the dichloromethane solution of compound 7 and compound 5, pyridine is added;
(viii) pH to about 9.0 of the intermediate that regulation separates, to produce compound 8;
(ix) to the mixing of the cytosine in anhydrous N,N-dimethylformamide He (S-) trityl glycidyl ether Thing adds potassium carbonate, and is heated to about 90 DEG C, to provide compound 9 (with raceme or enantiopure form);
X the mixture of () compound 8 in DMF and compound 9 adds tert-butyl alcohol magnesium, to produce compound 10;
(xi) compound 10 is processed, to produce compound 11 with HCl (methanol or the succedaneum of organic solvent);
(xii) in compound 11, add water, and be heated to about 90-120 DEG C, to provide compound 12.
The invention still further relates to a kind of be used in treatment or prophylaxis of viral infections or the disease relevant with virus infection or disease, example As, the pharmaceutical preparation of the compound of the present invention in the method that double-stranded DNA (dsDNA) virus infects.
The pharmaceutical preparation that the invention still further relates to the present invention for treatment or prophylaxis of viral infections and/or is having with virus infection Application in the preparation of the medicine of the disease closed or disease (such as, dsDNA virus infects).
The invention still further relates to for treatment or prophylaxis of viral infections and/or the disease relevant with virus infection or disease (example As, dsDNA virus infects) method.
Except as otherwise noted, all of scientific and technical terminology used herein is with those skilled in the art's The conventional implication understood is identical.In the case of a conflict, according to the description of the present invention, including definition.In the description, odd number Form also includes plural number, unless the context.Although described herein, those are similar or be equivalent to the present invention Method or material can the practice of the present invention or test in use, but be suitable for method and material hereinafter retouched State.All publications, patent application, patent and other list of references mentioned by Ben Wen are incorporated herein by.Draw herein List of references do not allow the prior art for the invention being claimed.Additionally, material, method and embodiment only rise illustrative Effect, it is not intended that limit.
Detailed description below and claims will clearly display other feature and advantage of the present invention.
Detailed description of the invention
The invention provides compound, pharmaceutical composition and synthetic method and use compound to treat or pre-diseases prevention Poison infects or the method for the disease relevant with virus infections or disease (such as, dsDNA virus infection).
Compared with the compound used similar with this area, the compound of the present invention has the curative effect/toxicity ratio of improvement.
Definition
For the purposes of the present invention, following definition will be used (except as otherwise noted):
Term " compound (odd number) of the present invention " or " compound (plural) of the present invention " refer to chemical combination disclosed herein Thing, such as, the compound of the present invention includes any compound in the compound shown in formula disclosed herein (I).Whenever this When term uses in the context of the present invention, it should be understood that be referred to free alkali and its pharmaceutically acceptable salt, if In these cases, it is so possible and/or suitable.Should be appreciated that compound described herein 12-24 is the change of formula (I) The subset of compound.
Term as used herein " alkyl " refers to saturated, the alkyl of straight or branched, in certain embodiments, wraps respectively Include the carbon atom between to six or to eight.It is one or more rudimentary that side chain refers to be connected in linear alkyl chain C1-C6Alkyl, such as methyl, ethyl or propyl group.Typical alkyl includes methyl, ethyl, n-pro-pyl, isopropyl, normal-butyl, uncle Butyl, n-pentyl and 3-amyl group.C1-C6The example of alkyl include but not limited to methyl, ethyl, propyl group, isopropyl, butyl, uncle Butyl, neopentyl, n-hexyl;C1-C8The example of alkyl include but not limited to methyl, ethyl, propyl group, isopropyl, normal-butyl, The tert-butyl group, neopentyl, n-hexyl, heptyl, octyl group.
Term as used herein " thiazolinyl " represents from the monovalence with at least one carbon-carbon double bond that hydrocarbonaceous part is derivative Group, in certain embodiments, containing from two to six or two to eight carbon atoms.Double bond may yes or no another The junction point of group.C2-C8The example of thiazolinyl includes but not limited to, such as, and vinyl, acrylic, cyclobutenyl, 1-methyl-2-fourth Alkene-1-base, heptenyl, octenyl etc..
Term " alkoxyl " refers to-O-alkyl.
Term as used herein " aryl " refer to have one or more aromatic ring, condense or the monocycle of non-condensed or multi-ring Carbon-loop system, include but not limited to, phenyl, naphthyl, tetralyl, indanyl, indenyl etc..Term aryl includes dihydro Yin Diindyl.
Term as used herein " cycloalkyl " represents by monocycle or many ring fillings or part undersaturated carbocyclic ring chemical combination The univalent perssad that thing is derivative.C3-C8The example of cycloalkyl (3 to 8 yuan of cycloalkyl) include but not limited to, cyclopropyl, cyclobutyl, Cyclopenta, cyclohexyl, cyclopenta and ring octyl group;C3-C12The example of cycloalkyl include but not limited to, cyclopropyl, cyclobutyl, ring Amyl group, cyclohexyl, bicyclo-[2.2.1] heptyl and dicyclo [2.2.2] octyl group.Involved can also be from having at least one carbon The monocycle of carbon double bond or multi-ring carbocyclic compound are by removing the derivative univalent perssad of a hydrogen atom.The example of these groups Son includes but not limited to, cyclopropanyl, cyclobutane base, cyclopentenyl, cyclohexenyl group, cycloheptenyl, cyclo-octene base etc..
Term as used herein " heteroaryl " refer to have at least one aromatic ring, monocycle or multi-ring (such as, dicyclo or Three rings or multi-ring) condense or the group of non-condensed or loop systems, described aromatic ring has 5-10 annular atoms, and one of them ring Atom is selected from S, O and N, and zero, one or two annular atoms are other hetero atoms being independently selected from S, O and N.
Term " 5 or 6 yuan of heteroaryls " refers to have five or six annular atomses and one of them annular atoms selected from S, O and N Ring.Heteroaryl includes but not limited to, pyridine radicals, pyrazinyl, pyrimidine radicals, pyrrole radicals, pyrazolyl, imidazole radicals, thiazolyl, oxazole Base, isoxazolyl, thiadiazolyl group, oxadiazoles base, thienyl, furyl, quinolyl, isoquinolyl, benzimidazolyl, dibenzo Oxazolyl, quinoxalinyl etc..
Term as used herein " 3-8 unit heterocycle " refers to non-aromatic 3,4,5,6,7 or 8 ring or the dicyclo that condenses or The non-condensed system of three rings, wherein (i) each ring comprises one to three hetero atom independently selected from O, S and N;(ii) each 5 Ring has 0-1 double bond and each 6 rings to have 0-2 double bond;(iii) described N and S hetero atom is the most oxidized; (iv) described atom N can be the most quaternized, and (iv) any of above ring can condense with phenyl ring.Typical heterocyclic group includes But it is not limited to, [1,3] dioxolanes, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidyl, piperazine Piperazine base, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolinyl, isothiazoline base and tetrahydrofuran base.
According to the present invention, any aryl described herein, substituted aryl, heteroaryl and substituted heteroaryl can be Arbitrarily aromatic group.Aromatic group can be substituted or unsubstituted.
Term used herein " halogen " and " halogen " refer to the atom selected from fluorine, chlorine, bromine and iodine.
Substituted or unsubstituted: as described herein, the compound of the present invention is optionally taken by one or more substituent groups In generation, such as generally by explained above, or by the specific classification of the present invention, the kind illustration of subclass sum.It should be appreciated that Phrase " optionally substituted " uses phrase " substituted or unsubstituted " convertibly.It is said that in general, term " substituted ", no matter Above with or without term " optionally ", all represent replacing to the hydrogen-based in fixed structure at the group with specified substituent.Unless Being otherwise noted, optionally substituted group can have substituent group in each commutable position of this group, and ought arbitrarily give The more than one substituent group that in fixed structure, more than one position can be selected from special groups is replaced, taking of each position Can be same or different for base.
When this paper term " medicine " or " pharmaceutically acceptable " refer to when using as adjective that for receiver be base This is nontoxic and the most harmless.Phrase used herein " pharmaceutically acceptable " refers in the range of rational medical judgment It is applicable to there is no excessive toxicity, stimulation, anaphylaxis or other problems or complication with the contact tissue of human and animal, Those compounds, material, compositions, carrier and/or the dosage form suitable with rational interests/Hazard ratio.
" pharmaceutical preparation " also refers to that carrier, solvent, excipient and salt must be with the active component (changes of the such as present invention of preparation Compound) compatible.One of ordinary skill in the art will appreciate that term " pharmaceutical preparation " and " pharmaceutical composition " are the most interchangeable , they thus be accordingly used in the purpose of the application, and includes being suitable for administering to mammal, the preparation of the such as mankind.
" pharmaceutical composition " used herein relates to comprising to be suitable for administering to the present invention's of the form of experimenter The preparation of compound.In one embodiment, pharmaceutical composition is in bulk or unit dosage forms.Described unit dosage forms is any Various forms, including, the such as single pump of capsule, IV bag, tablet, aerosol inhaler or bottle.Chemical combination in unit dose The amount of the active component (such as, compound disclosed in preparation or salt, hydrate, solvate or its isomer) in thing is effective Measure and change according to involved particular treatment.One skilled in the art is it will be appreciated that sometimes for according to patient Age and situation dosage is made routine change.Dosage also depends on route of administration.There is number of ways it is contemplated that include mouth In clothes, pulmonary, rectum, parenteral, transdermal, subcutaneous, intravenous, muscle, intraperitoneal, suction, oral cavity, Sublingual, pleura, in sheath, Intranasal etc..For the local of compound of the present invention or applied dermally dosage form include powder, spray, ointment, paste, cream, Lotion, gel, solution, patch and inhalant.In one embodiment, reactive compound and pharmaceutically acceptable carrier are in nothing Mix under the conditions of bacterium, and by required any preservative, buffer agent or propellants.
" pharmaceutically acceptable carrier " used herein can include any and all of solvent, diluent or Other liquid vehicle, dispersion or suspension aids, surfactant, isotonic agent, thickening agent or emulsifying agent, preservative, solid glue Mixture, lubricant etc., as being applicable to required particular dosage form.Remington’s Pharmaceutical Sciences, Sixteenth Edition, E.W.Martin (Mack Publishing Co., Easton, Pa., 1980) disclose for joining The various carriers of pharmacy compositions and technology of preparing known to it.Unless it is incompatible with compound at any conventional carrier medium In the case of, such as by producing any less desirable biological effect or other any with harmful way and pharmaceutical composition Other component interacts, and its use is contained within the scope of the present invention.Can be as the material of pharmaceutically acceptable carrier The example of material includes but not limited to, sugar, such as lactose, dextrose plus saccharose;Starch, such as corn starch and potato starch; Cellulose and its derivates, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate;Powdered tragacanth;Fructus Hordei Germinatus;Bright Glue;Talcum;Excipient, such as cocoa butter and suppository wax;Oil, such as Oleum Arachidis hypogaeae semen, Oleum Gossypii semen, safflower oil, Oleum sesami, olive oil, jade Miyou and soybean oil;Glycol, such as propylene glycol;Ester, such as ethyl oleate and ethyl laurate;Agar;Buffer agent, such as magnesium hydroxide and Aluminium hydroxide;Alginic acid;Apirogen water;Isotonic saline solution;Ringer's mixture;Ethanol and phosphate buffered solution, and other non-poison The lubricant that property is compatible, such as sodium lauryl sulfate and magnesium stearate, and coloring agent, releasing agent, coating materials, sweeting agent, seasoning Agent and aromatic, preservative and antioxidant can also be present in compositions, according to the judgement of formulator." pharmaceutically Acceptable excipient or carrier " further relate to a kind of for prepare universal security, nontoxic and the most biologically or its The pharmaceutically acceptable excipient of the less desirable pharmaceutical composition of its aspect or carrier, and include can be by veterinary purpose and people The excipient that class medicinal usage accepts.Such as " pharmaceutically acceptable excipient " bag used in the specification and in the claims Include a kind of and more than one such excipient.
Some compounds of the present invention can be presented in non-solvated and solvation, such as, and hydrate.
" solvate " refers to the solvent addition form of a kind of solvent comprising stoichiometric or non-stoichiometric amount.One A little compounds have the trend of a kind of solvent molecule forming fixed molar ratio at crystalline solid state, thus form solvate. If solvent is water, then the solvate formed is hydrate, and when solvent is alcohol, the solvate of formation is alcoholates.Water Compound is by remaining in that its molecularity such as H by one or more water wherein2The material of O combines and is formed, this combination One or more hydrates can be formed.In hydrate, hydrone is contacted, particularly by the secondary valency of intermolecular force Hydrogen bond.Solid hydrate comprises the water as so-called water of crystallization of stoichiometric proportion, and wherein hydrone is relative to their knot Conjunction state needs not to be equivalence.The example of hydrate is a times semihydrate, monohydrate, dihydrate or trihydrate.This The hydrate of the salt of bright compound is similarly suitable.
Present invention additionally comprises the metabolite of compound as herein described.No matter it is intrinsic or pharmacy, from chemicalization The metabolite of compound is as the part formation of degraded and the native biochemical process eliminating compound.The degradation rate of compound is The persistent period of its effect and the important determiner of intensity.Analyze the metabolite of pharmaceutical composition, drug metabolism is that medicine is opened The important component part sent out so that the understanding to any adverse side effect.
Physiologically/pharmaceutically acceptable/compatible salt: physiologically acceptable, i.e. pharmaceutically compatible, salt can To be the salt of the compound with mineral acid or organic acid of the present invention.Preferably mineral acid, e.g., such as, hydrochloric acid, hydrogen bromine Acid, phosphoric acid or the salt of sulphuric acid, or organic carboxyl acid or sulfonic acid, e.g., such as, acetic acid, trifluoroacetic acid, propanoate hydrochloride, maleic acid, rich horse Acid, malic acid, citric acid, tartaric acid, lactic acid, benzoic acid or methanesulfonic acid, ethyl sulfonic acid, benzenesulfonic acid, toluenesulfonic acid or naphthalenedisulfonic acid Salt.
The salt of other pharmaceutically compatible that can be mentioned that is the salt of conventional alkali, e.g., such as, alkali metal salt (such as sodium salt or Potassium salt), alkali salt (such as calcium salt or magnesium salt) or derive from salt ammonia or organic amine, e.g., such as, diethylamine, triethylamine, second Base diisopropylamine, procaine, dibenzylamine, N-methylmorpholine, dihydro Colophonium or the ammonium salt of methyl piperidine.
" pharmaceutically acceptable salt " used herein refers to the derivant of the compound of the present invention, wherein, parent chemical combination Thing comes modified by preparing its acid or alkali salt.The example of pharmaceutically acceptable salt includes but not limited to, alkaline residue such as amine Mineral or acylate, the alkali metal salt of acidic residues such as carboxylic acid or organic salt etc..Pharmaceutically acceptable salt includes routine The quaternary ammonium salt that non-toxic salt or parent compound are formed, such as, from non-toxic inorganic or organic acid.Such as, these conventional nothings Poison salt include but not limited to those from selected from Aspirin, 2-ethylenehydrinsulfonic acid, acetic acid, ascorbic acid, benzenesulfonic acid, Benzoic acid, heavy carbonic, carbonic acid, citric acid, ethylenediaminetetraacetic acid, ethane disulfonic acid, 1,2-ethane sulfonic acid, fumaric acid, glucoheptose Acid, gluconic acid, glutamic acid, glycolic, p-aminophenyl glycolic, hexylresorcin, hydroxamic acid, hydrobromic acid, hydrochloric acid, hydrogen Iodic acid, hydroxymaleic acid, hydroxyl naphthalene, isethionic acid, lactic acid, lactobionic acid, lauryl sulfonic acid, maleic acid, malic acid, mandelic acid, Loprazolam, octanoic acid, nitric acid, oxalic acid, flutter acid, pantothenic acid, phenylacetic acid, phosphoric acid, polygalacturonic acid, propanoic acid, salicylic acid, tristearin Acetic acid sour, sub-, succinic acid, sulfamic acid, p-aminobenzene sulfonic acid, sulphuric acid, tannic acid, tartaric acid, toluenesulfonic acid and generally occur within Amino acid, such as, mineral acid derivative in glycine, alanine, phenylalanine, arginine etc. and organic acid.
Other example of pharmaceutically acceptable salt includes caproic acid, Pentamethylene. propanoic acid, acetone acid, malonic acid, 3-(4-hydroxyl Benzoyl) benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2-LOMAR PWA EINECS 246-676-2,4-toluenesulfonic acid, camphorsulfonic acid, 4-methyl bicyclic [2.2.2]-oct-2-ene-1-carboxylic acid, 3-phenylpropionic acid, trimethylace tonitric, butylacetic acid, muconic acid etc..Present invention additionally comprises When the acid proton being present in parent compound or by metal ion, such as alkali metal ion or alkaline-earth metal ions, example As, aluminium ion, replace the salt formed;Or with organic base such as ethanolamine, diethanolamine, triethanolamine, trometamol, N-methyl Portugal Osamine, diethylamine, DEAE diethylaminoethanol, ethylenediamine, imidazoles, lysine, arginine, morpholine, 2-hydroxyethyl morpholine, dibenzyl The salt that ethylenediamine, trimethylamine, piperidines, pyrrolidine, benzylamine, tetramethyl oxyammonia etc. cooperatively form.
Term as used herein " is treated " and is referred to subtract with any suitable degree in the patient currently having this situation Few symptom, mark, and/or the situation of any side effect.In certain embodiments, treatment can be administered to those and only opens up Reveal the experimenter of the early stage sign of this situation, it is therefore an objective to reduce disease, disease and/or the risk of situation development.
Term as used herein " prevents " refer to any part ground or fully prevent or delay disease, disease and/or disease One or more symptoms of feelings or the method for the outbreak of feature.Prophylactic treatment can be administered to those do not demonstrate disease, The experimenter of the sign of disease and/or the state of an illness.
Term as used herein " therapeutically effective amount " refers to treatment, improves or prevents disease or the state of an illness identified, or aobvious The amount of the medicament of detectable treatment or inhibition is shown.This effect can pass through any detection side as known in the art Method detects.The accurate effective dose of experimenter is depended on the body weight of experimenter, height and health status;The essence of the state of an illness and disease The degree of feelings;And select medicine or the combination of medicine used.When given, therapeutically effective amount can be led to Crossing normal experiment to determine, this experiment is in the technical ability and determination range of clinicist.
" experimenter " used herein refers to the mankind or animal (in the case of animal, more generally mammal).? One aspect, experimenter is the mankind.In one aspect, experimenter is man.In one aspect, experimenter is woman.
It should be appreciated that all quote pharmaceutically acceptable salt includes the most identical salt Solvent addition form (solute) or crystal form (polymorph).
The compound of the present invention can also be prepared to ester, such as, pharmaceutically acceptable ester.Such as, in compound Carboxylic acid functional can change into its corresponding ester, such as, methyl ester, ethyl ester or other ester.Additionally, the hydroxyl in compound is permissible It is converted into its corresponding ester, such as, acetas, propionic ester or other ester.
United States Patent (USP) 7749983, during entire contents is incorporated herein by reference by entirety, relates to phosphonate ester, nucleoside phosphonate Ester or nucleoside phosphonate compound, including end or side chain second from the bottom, the undersaturated and alkoxyalkyl phosphine of halogen substiuted Ester compound.In some other embodiments, the compound of formula (I) may be embodied in United States Patent (USP) 7749983 as R base The disclosed side chain of group.
The present invention includes generally by the noval chemical compound shown in formula (I), or its pharmaceutically acceptable salt, and its preparation method With application.
The compound of the present invention can also prepare prodrug.In certain embodiments, one or more of the present invention are changed Compound is formulated into prodrug.In certain embodiments, when using in vivo, prodrug was chemically converted as biology Upper, pharmaceutically or form more active in treatment.In certain embodiments, prodrug is useful, because its corresponding Activity form is compared, and they are easier to use.Such as, in some example, its corresponding activity form of prodrug is compared Possible bioavailability higher (such as, by Orally administered).In certain embodiments, prodrug its corresponding activity shape Formula compares the dissolubility being likely to be of improvement.In certain embodiments, the water solublity of prodrug activity form more corresponding than it Less.In certain embodiments, these prodrugs have the cross-cell membrane transmission of excellence, and in these cell membrane, water solublity is not It is beneficial to flowing.In certain embodiments, prodrug is ester.In some such embodiment, ester is metabolism hydrolysis when using For carboxylic acid.In certain embodiments, the compound containing carboxylic acid is corresponding activity form.In certain embodiments, prodrug Including the small peptide (polyamino acid) being connected to acidic group.In some such embodiment, it is corresponding that peptide cracks formation when using Activity form.
In certain embodiments, prodrug is produced by modified reactive compound pharmaceutically so that when executing in vivo Used time can produce reactive compound.Prodrug can be designed as changing drug metabolism stability or transport property, covers pair Effect or toxicity, improve the taste of medicine or change other characteristics of medicine.By well known to a person skilled in the art internal medicine Effect process and drug metabolism, once pharmaceutically active compound is known, it is possible to design the prodrug (example of this compound As, see Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press,New York,pages 388-392)。
Compound or its pharmaceutically acceptable salt, ester or derivant are by oral, per nasal, percutaneous, lung, suction, cheek, tongue Under, intraperitoneal, in subcutaneous, intramuscular, intravenous, rectum, pleura, in sheath and parenteral administration.In one embodiment, this chemical combination Thing is Orally administered.It would be recognized by those skilled in the art that the advantage of some route of administration.
The dosage regimen utilizing compound is including type, species, age, body weight, sex according to the many factors of patient With medical conditions, the order of severity of the state of an illness to be treated, route of administration, the kidney of patient and liver function and its use specific Compound or its salt selects.The doctor of one ordinary skill or veterinary can determine easily and output the required of effective dose Medicine prevents, resists or stops the development of the state of an illness.
The compound of the present invention
Generally, the nucleotide phosphate of the present invention can also be represented by such as following formula (I):
Wherein, R is:
The typical nucleotide phosphate of the present invention is listed in table 1.
Table 1
In a further embodiment, the invention provides there is formula Compound 8 and the method for synthesis compound 8, wherein, described method comprises the steps:
I () adds magnesium chips in 2-methyltetrahydrofuran (Me-THF) solution of bromo-3 methybutanes of 1-, be subsequently heated, so This mixture of rear cooling;
(ii) in the reactant mixture of step (i), the Me-THF solution of the bromo-DODECANOL, 1-of 12-is added, immediately after Add oxolane (THF) solution of four chloro copper acid two lithiums;
(iii) with the aqueous phase in ethyl acetate back extraction reactant mixture;
(iv) by saline washing organic solution and through MgSO4It is dried, filters, concentrate the most in a vacuum, to produce chemical combination Thing 3;
V () adds methylsulfonyl in compound 3 and N, the cold soln of the dichloromethane of N-diisopropylethylamine (DIPEA) Chlorine, maintains the temperature at less than about 5 DEG C simultaneously;
(vi) reaction is warmed to room temperature, and stir several hours;
(vii) in reactant mixture, add mesyl chloride and DIPEA, and stir several hours;
(viii) add water while cooling reactant mixture, from water layer, then separate dichloromethane (DCM) layer, through dry Drying prescription is dried, and filters DCM layer to remove desiccant;
(ix) DCM solution it is concentrated under vacuum, to provide yellow oil;And add in the concentrate of yellow oil Methanol;
X () filters the methanol concentrate being settled out white solid, cross filter solid and be dried, and prepares compound 4;
(xi) in the cold soln of the METHYLPYRROLIDONE (NMP) of 1,3-PD, sodium hydride (NaH) is added, and Warm this mixture;
(xii) in the solution of step (xi), add compound 4, and stir several hours;
(xiii) in solution, add water and ethyl acetate, and separate organic layer;
(xiv) concentration of organic layers in a vacuum, adds methanol, is then dried;
(xv) add acetonitrile, repeat step (xiv), after filtration and drying, form compound 5;
(xvi) in the acetonitrile solution of compound 6 (purchased from Lacamas laboratory), trimethyl alkane silicon bromine (TMS-is added Br);
(xvii) add dichloromethane after removing acetonitrile and TMS-Br, then concentrate;
(xviii) add oxalyl chloride and dimethylformamide (DMF), and concentrate in a vacuum, to form compound 7;
(xix) mixing cpd 5 and compound 7 in dichloromethane, and add in the solution of compound 7 and compound 5 Enter pyridine;
(xx) organic layer is separated after adding water in the mixture to step (xix);
(xxi) organic layer is again separated after adding water and methanol in the organic layer to step (xx);
(xxii) it is dried in a vacuum the organic layer from step (xxi), and adds acetone;
(xxiii) it is dried and before being further dried, adds acetone and regulate pH to about 9.0;
(xxiv) acetone is added after filtering the solid formed in step (xxiii);With
(xxv) solid product of also drying steps (xxiv) is filtered, to produce compound 8.
In another embodiment, described method comprises the steps:
I () adds in 2-methyltetrahydrofuran (Me-THF) solution of bromo-3 methybutanes of 1-(or suitably replacing hydrocarbon) Magnesium chips;
(ii) in reactant mixture, add the Me-THF solution of the bromo-DODECANOL, 1-of 12-(or suitably replacing hydrocarbon), with Oxolane (THF) solution of rear addition four chloro copper acid two lithiums, to synthesize compound 3;
(iii) in compound 3 and N, the dichloromethane cold soln of N-diisopropylethylamine (DIPEA), methylsulfonyl is added Chlorine, maintains the temperature at less than about 5 DEG C simultaneously, to provide compound 4;
(iv) in the cold soln of the METHYLPYRROLIDONE (NMP) of 1,3-PD, sodium hydride (NaH) is added, with Rear addition compound 4, to produce compound 5;
V () adds bromotrimethylsilane (TMS-Br) in the acetonitrile solution of compound 6 (purchased from Lacamas laboratory);
(vi) oxalyl chloride and dimethylformamide (DMF) are added, to form compound 7 after removing acetonitrile and TMS-Br;
(vii) in the dichloromethane solution of compound 7 and compound 5, pyridine is added;
(viii) pH to about 9.0 of the intermediate that regulation separates, to produce compound 8.
The compound 12-24 synthesized according to the method for the present invention there is no impurity.Synthesize according to the embodiment of the present invention Compound 12-24 there is the purity greater than or equal to about 99%w/w.Should be appreciated that method disclosed by the invention can be suitable for In the extensive of required compound and preparation on a small scale.In the preferred embodiment of approach described herein, phosphonate ester can With by large-scale production, such as, on an industrial scale rather than tentative/laboratory scale.Such as, the method according to the invention At least 1g, or at least 5g, or at least 10g, or at least 100g, or at least 1kg are prepared in one batch process permission, or at least The batch of the phosphonate product of 100kg.The compound of the present invention can be made into enantiomer, diastereomer and racemic modification.Additionally, The method allows to have the preparation of phosphonate product of purity of recorded at least 98% or at least 98.5% by HPLC.At this In invention preferred embodiment, these products be not related to by any type of chromatography (such as, gas chromatogram, HPLC, system Standby liquid chromatograph, size exclusion chromatograph etc.) purification reaction sequence in obtain.
In one embodiment, the present invention relates to compound 12:
In one embodiment, the present invention relates to compound 13:
In another embodiment, the one or more-CH on the side chain of compound 12 and/or compound 132Group In one or two hydrogen optionally replaced by alkyl, halogen or other group any as disclosed in US 7749983.
In certain embodiments, end group-CH3Taken by halogen or alkyl or other group any as disclosed in US 7749983 Generation.Such as, one embodiment of the present of invention relates to compound 22, wherein, end group-CH3Group is by fluorine-based replacement:
Synthetic method
The invention provides the synthetic method of branched nucleosides phosphonate ester.In some aspects, the invention provides generally by formula (I) preparation method of the compound shown in:
Wherein, R is:
In one embodiment, the invention provides the preparation method of the typical compound being listed in table 1.
The embodiment provides the compound of formula (I) or its pharmaceutically acceptable salt or the synthesis of solvate Method.In one embodiment, the invention provides a kind of compound 12 and/or the synthetic method of compound 13.The present invention is also Provide the synthetic method of a kind of compound 8.
The invention provides the synthetic method of substituted phosphonate ester.In some aspects, the invention provides there is structure:
The preparation method of compound 12.
In one embodiment, the invention provides there is structure:
Compound 13, or it is right Reflect body, diastereomer, racemic modification or its mixture, or the preparation method of pharmaceutically acceptable salt.
The invention provides midbody compound 8 and its synthetic method.
Following scheme and explanation describe some preparation methoies of the compound of the present invention.
The synthetic method of the present invention can allow various functional group;Therefore various substituted initial material can be used Material.Described method generally or close to providing required finalization compound at the end of whole process, although wishing in some cases Hope and finalization compound is further converted into its pharmaceutically acceptable salt, ester or derivant.
Scheme 1
The synthesis of compound 3: by solvent (such as, the 2-methyltetrahydrofuran (Me-of 1-bromo-3-methybutane THF) solution) adds the compound 3 of alkaline-earth metal (such as considering the magnesium of form to be worth doing) the synthesis present invention.In order to prevent temperature liter Height, uses dry ice and ketone (such as, acetone) bath.At about 30-60 DEG C, such as about 40 DEG C, add halogen (such as a, fritter Iodine).Then solution is heated to about 60-80 DEG C (such as, about 61 DEG C), and stir about 1-3 hour is (such as, about 1.1,1.2, 1.3,1.4,1.5,1.6,1.7,1.8,1.9,2.0,2.1,2.2,2.3,2.4,2.5,2.6,2.7,2.8,2.9 or 3.0 little Time).Mixture is cooled to about 30-60 DEG C (such as, equal to or less than about 40 DEG C).Reactant mixture is cooled further to about 0 to-70 DEG C (such as, about-59 DEG C).The organic of alcohol (such as, the bromo-DODECANOL, 1-of fatty alcohol such as 12-) is added in mixture Solvent (such as, Me-THF).After adding alcohol (such as, fatty alcohol such as 12 bromo-DODECANOL, 1-s), add catalyst solution immediately (such as, the four sour two lithium solution of chloro copper (II)).Reaction is warmed to about room temperature stir several hours (such as, about 16 hours). After completion of the reaction, reaction is cooled to about 0 DEG C, and inorganic solvent (such as ammonium chloride) is slowly added to until saturated and straight It is increased to about 15-30 DEG C (such as, greater than about 22 DEG C) to temperature.Aqueous phase organic additive (such as, ethyl acetate) back extraction. The Organic substance merged with saline solution (such as, saline) washing, through inorganic salt (such as, MgSO4) be dried, then filter.About 40 DEG C vacuum in concentrate this solution.
The synthesis of compound 4: but to compound 3 be that (such as, N, N-bis-is different for the organic reagent of highly basic weak nucleophilic reagent Propylethylamine (DIPEA)) solvent (such as, dichloromethane) cold soln in be slowly added to mesyl chloride, to guarantee that temperature is not More than about 5 DEG C can be raised to.Methylsulfonyl agent (such as, mesyl chloride (methyl is added in the reactant mixture containing compound 3 Sulfonic acid chloride) and DIPEA.After cooling down this mixture, add water and stir.Dichloromethane (DCM) layer is separated, through Na2SO4Dry Dry, and filter.This solution is concentrated in about 40-50 DEG C vacuum.Methanol is added in yellow residue.By this solution at about 4 DEG C Lower standing about 30min is to be settled out white solid.Filter the solution of precipitation solid, and solid is air-dried on the filter several little Time.Concentrate the filtrate to half volume and filter.Solid is merged, and grinds in methanol.Filter and dry white solid.
The synthesis of compound 5: add suitably in the cold soln of the 1,3-PD less than 0 DEG C (such as, about-5 DEG C) Organic reagent (such as, METHYLPYRROLIDONE (NMP)) and highly basic (such as NaH).Mixture is warming to room temperature, and stirs Mix about 30min.The compound 4 being dissolved in suitable organic reagent (such as NMP) joins in this solution.To reaction mixing Thing adds water and organic solvent (such as ethyl acetate).Organic layer is separated with water layer.Wash organic layer with water.True at 40 DEG C Aerial this solution of concentration.This mixture is further dried by adding methanol and concentrates in about 40 DEG C of vacuum.By the non-matter of polarity Sub-solvent (such as acetonitrile) repeats the step being dried in methanol and concentrating.Polar non-solute is added in yellow oil (such as acetonitrile), and stir this mixture.Form wax-like white solid, filtered.It is dried to obtain in a rotary evaporator Compound 5.
Scheme 2
The synthesis of compound 8: add front three bromide in the solution of the polar non-solute (such as acetonitrile) of compound 6 Silane (TMS-Br).After having added, internal temperature is regulated to about 55 DEG C.After stirring the mixture for 2 hours, by about 40 Polar non-solute (such as acetonitrile) and TMS-Br are removed by the vacuum distilling at DEG C, form concentrate.Add in concentrate Enter organic solvent (such as dichloromethane), to form solution, be subsequently added suitable organic reagent (such as oxalyl chloride).Organic After the addition of reagent (such as oxalyl chloride) completes, add organic solvent (such as polarity (hydrophilic) aprotic solvent (such as, two Methylformamide (DMF)).Stirring reactant mixture, then concentrates in the vacuum under the external temperature of about 35 DEG C, with offerization Compound 7.Intermediate compound 7 can be used for next step without purification.
(e.g., from about-8 DEG C) cooling compound 5 and the dichloromethane of compound 7 (0.1462mol, 44.33g) below 0 DEG C (423ml) solution.Pyridine (0.381mol, 30.18g) is added in the solution of this cooling.Reactant mixture is stirred 3 hours. Carrying out thin layer chromatography (TLC) with the volume ratio of hexane Yu ethyl acetate for 2:1, its display compound 5 is consumed.To reaction Mixture (being cooled to 10 DEG C) adds 200ml water.This mixture is stirred 0.5 hour.It is then peeled off organic layer.To organic layer Middle addition 100ml water and 75ml methanol.Again separate organic layer, and concentrate in the vacuum of 35 DEG C.Add in residue 200ml acetone, and use 6N NaOH (about using 15ml) by pH regulator to about 9.04.This mixture is stood at about 4 DEG C 16 Hour.After incubation period, 10g white solid precipitates.The mixture precipitated containing white solid is filtered.Add in this mixture Enter other 300ml acetone.Mixture is stood 16 hours again at 4 DEG C.After other incubation period, sepia solid sinks Form sediment.Filter and be dried this mixture, to produce compound 8.
Scheme 3
The synthesis of compound 10: by compound 9, compound 8, gentle alkaline-earth alkoxides (alkali), (the such as tert-butyl alcohol Magnesium) it is heated to about 60 DEG C or higher temperature (such as 80 DEG C) together with DMF (25ml), it is lasting that within more than 1 hour, (e.g., from about 3 is little Time).This reaction is cooled in organic solvent (such as isopropyl acetate) room temperature, and adds acid (such as HCl).By organic layer Separate, wash organic layer with saline solution (such as saline), through salt (such as Na2SO4) be dried, and concentrate in about 40 DEG C of vacuum.To This mixture adds methanol, concentrates to produce compound 10.
Scheme 3a
The synthesis of compound 10a: by compound 9a, compound 8, gentle alkaline-earth alkoxides (alkali), (the such as tert-butyl alcohol Magnesium) it is heated to about 60 DEG C or higher temperature (such as 80 DEG C) together with DMF (25ml), continue more than 1 hour, e.g., from about 3 hours. By this reaction at organic solvent, such as isopropyl acetate is cooled to room temperature, and adds acid (such as HCl).Organic layer is separated, Organic layer is washed, through salt (such as Na with saline solution (such as saline)2SO4) be dried, and concentrate in about 40 DEG C of vacuum.To mixing Thing adds methanol, concentrates to produce compound 10a.
Scheme 4
The synthesis of compound 11: at room temperature add alcohol (such as methanol) and acid (such as HCl) in compound 10.Formed And leach white solid trityl by-product.Dilute with water filtrate, and use alkali metal hydroxide (such as NaOH) to regulate PH to about 2.5, forms compound 11.By product at organic solvent, such as acetone is made slurry, and filters.By product in room temperature Under vacuum drying oven in be dried.
Scheme 4a
The synthesis of compound 11a: at room temperature add alcohol (such as methanol) and acid (such as HCl) in compound 10a.Shape Become and leach white solid trityl by-product.Dilute with water filtrate, and use alkali metal hydroxide (such as NaOH) to adjust Joint pH to about 2.5, forms compound 11a.By product at organic solvent, such as acetone is made slurry, and filters.By product in room Vacuum drying oven under Wen is dried.
Scheme 5
The synthesis of compound 12: add water in heating the mixture to the forward direction compound 11 of about 90 DEG C, and stir.? After having reacted 95% or more than 95%, mixture is cooled to room temperature.Use acid (such as HCl) by pH regulator to about 1-2.With Ethyl acetate extraction water solution.By the acetic acid ethyl ester extract of merging through Na2SO4It is dried, filters, and in the vacuum of about 40 DEG C Concentrate, to produce thick grease.Being ground by the grease that this is thick with acetone, cooling, to produce white solid.Filter and be dried White solid, to produce compound 12.
Scheme 5a
The synthesis of compound 13: add water in the forward direction compound 11a heat the mixture to about 90 DEG C, and stir. After reaction completes 95% or more than 95%, mixture is cooled to room temperature.Use acid (such as HCl) by pH regulator to about 1-2. It is extracted with ethyl acetate aqueous solution.By the acetic acid ethyl ester extract of merging through Na2SO4It is dried, filters, and in the vacuum of about 40 DEG C Middle concentration, to synthesize thick grease.With acetone, the grease that this is the thickest is ground, and cool down to produce white solid.Filter And dry white solid, to produce compound 13.
Similar method is used for preparing other compounds of formula (I).Initiation material, such as compound 1 and/or compound 3 can need according to the synthesis of the compound of the present invention and change.
Throughout the specification, wherein, method or process are described as having, include or comprise and specifically process step, Described process also basically comprises or is made up of described process step.Moreover, it should be understood that the order of step or perform some The order of action is unessential, as long as the present invention keeps operability.Additionally, two or more steps or action can be simultaneously Carry out.
The purity of compound
The invention provides the compound of formula (I), such as, have greater than or equal to about 91%w/w, greater than or equal to about In 95%w/w, or the compound 12-24 (or its pharmaceutically acceptable salt) of the purity greater than or equal to about 99%w/w arbitrarily A kind of.
In certain embodiments, the compound shown in formula (I) of the present invention, such as, compound 12-24 there is no miscellaneous Matter.In certain embodiments, the purity of compound 12-24 or its pharmaceutically acceptable salt equal to or more than 92% (such as, >= 92%, >=93%, >=94%, >=95%, >=96%, >=97%, >=98%, >=99%, or >=99.5%).Implement at other In example, the purity of compound 12-24 or its pharmaceutically acceptable salt equal to or more than 91% (such as, >=91%, >=92%, >=93%, >=94%, >=95%, >=96%, >=97%, >=98%, >=99%, or >=99.5%).In another embodiment In, compound 12-24 or its pharmaceutically acceptable salt are solvates, such as, Methanol Solvate, alcohol solvent compound, Or isopropanol solvate.
In certain embodiments, the compound (such as compound 12-24) shown in formula (I) or its pharmaceutically acceptable salt Purity equal to or more than 92% (such as, >=92%, >=93%, >=94%, >=95%, >=96%, >=97%, >=98%, >= 99%, or >=99.5%).In other embodiments, any one in compound 12-24 (or its pharmaceutically acceptable salt) Have equal to or more than 91% (such as, >=91%, >=92%, >=93%, >=94%, >=95%, >=96%, >=97%, >= 98%, >=99%, or >=99.5%) purity.In one embodiment, (or it is pharmaceutically acceptable for compound 12-24 Salt) in any one purity be about 99%.
Present invention also offers and there is the purity equal to or more than 91%w/w, such as, have less than or equal to 9%w/w's Any one or its medicine in the compound (or its pharmaceutically acceptable salt) (such as, compound 12-24) of the formula (I) of impurity The therapeutic and/or preventative that on, acceptable salt infects in virus in experimenter (such as, immunodeficiency experimenter) Application in the preparation of the medicine for the treatment of.
The present invention provides the compound (such as compound 12-24) shown in formula (I) or its medicine of greater than about 99%w/w purity Acceptable salt on.In certain embodiments, any one in the compound (such as compound 12-24) shown in formula (I) Or the purity of its pharmaceutically acceptable salt is equal to or greater than about 99%w/w, 98%w/w, 97%w/w, 96%w/w, 95%w/ W, 94%w/w, 93%w/w, 92%w/w or 91%w/w.
Pharmaceutical composition
Throughout the specification, wherein, compositions is described as having, includes or comprise specific components, it is contemplated that Be described compositions the most substantially by or be made up of described component.
On the other hand, the invention provides the medicine group of the compound comprising formula (I) or its pharmaceutically acceptable salt Compound.In certain embodiments, the invention provides the medicine group of the compound comprising formula (I) or its pharmaceutically acceptable salt Compound and pharmaceutically acceptable carrier and/or diluent.Any in the pharmaceutical composition inclusion compound 12-24 of the present invention One or its pharmaceutically acceptable salt.Present invention also offers in inclusion compound 12-24 any one or its pharmaceutically Acceptable salt and a kind of pharmaceutically acceptable carrier and/or the pharmaceutical composition of diluent.
The preparation technology of the compounds of this invention and application process are shown in Remington:the Science and Practice of Pharmacy,19th edition,Mack Publishing Co.,Easton,PA(1995)。
In one embodiment, compound of the present invention and pharmaceutically acceptable salt thereof, with pharmaceutically acceptable Carrier or diluent combine be used in pharmaceutical preparation.Suitable pharmaceutically acceptable carrier includes inert solid filler Or diluent and sterilized water or organic solvent.Described compound will be present in such pharmaceutical composition, presents in an amount at least sufficient to provide Required dosage in scope of the present invention.
In another embodiment, the invention provides a kind of virus in experimenter (such as immunodeficiency experimenter) The therapeutic infected and/or preventative-therapeutic method, described method includes using purity equal to or more than 91% to experimenter In the compound 12-24 of w/w (such as, there is the impurity less than or equal to 9%w/w) any one or its pharmaceutically acceptable Salt.
(or it is pharmaceutically acceptable to the invention provides the compound of the formula (I) with the purity equal to or more than 91% Salt) (such as, in compound 12-24 any one or its pharmaceutically acceptable salt) in the virus infection for the treatment of experimenter Application in (infection of such as dsDNA virus).Described virus infects and includes but not limited to that human cytomegalovirus (HCMV), BK are sick Poison (BKV), epstein-Barr virus (EBV), adenovirus, JC virus (JCV), SV40, MC virus (MCV), KI virus (KIV), WU virus (WUV), vaccinia virus, herpes simplex virus type 1 (HSV-1), herpes simplex virus 2 (HSV-2), mankind's bleb Exanthema virus 6 type (HHV-6), human herpes virus type 8 (HHV-8), hepatitis B virus, hepatitis C virus, chickenpox banding bleb Exanthema virus (VZV), classical smallpox, alastrim, variola, cowpox, camel pox, monkeypox, poliovirus, Ebola virus, Marburg virus, enterovirus, papillomavirus and HIV (human immunodeficiency virus) (HIV) infect.Such as, described infection is right Valganciclovir hydrochlorate (or ganciclovir) has Drug resistance or demonstrates valganciclovir hydrochloric acid in described experimenter The side effect of salt (or ganciclovir).Or or it addition, there is the compound of the formula (I) of the purity equal to or more than 91%w/w (or its pharmaceutically acceptable salt), such as, any one or its pharmaceutically acceptable salt in compound 12-24, by with Treat CMV.Such as, human cytomegalovirus (HCMV).Such as, infect with after Therapeutic effect of ganciclovir, such as, this In the case of, cmv infection is acute.Described patient can be the patient of stem cell transplantation, such as, and the trouble of Bone Marrow Stem Cells Transplantation Person, is especially in the presence of the patient of the bone marrow toxicity risk (real or sense organ) of the ganciclovir come from patient.
In another embodiment, one in the compound of the formula (I) with the purity being equal to or greater than about 91% or Its pharmaceutically acceptable salt is administered orally in experimenter, and such as, about 0.01mg/kg is to about 10mg/kg or more, such as, The up to dosage of 100mg/kg.In another embodiment, there is the compound of the formula (I) of the purity being equal to or greater than about 91% Or its pharmaceutically acceptable salt is with about 0.01mg/kg, 0.05mg/kg, 0.1mg/kg, 0.5mg/kg, 1mg/kg, 1.5mg/ kg、2mg/kg、2.5mg/kg、3mg/kg、3.5mg/kg、4mg/kg、4.5mg/kg、5mg/kg、5.5mg/kg、6mg/kg、 6.5mg/kg, 7mg/kg, 7.5mg/kg, 8mg/kg, 8.5mg/kg, 9mg/kg, 9.5mg/kg or 10mg/kg or more dosage Or the dosage of any of which scope is administered orally in experimenter.
In certain embodiments, the compound (or its pharmaceutically acceptable salt) of formula (I), such as, the chemical combination of the present invention Any one in thing 12-24, with about 1-20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2- 1.3mg/kg, about 1.3-1.4mg/kg, about 1.4-1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7- 1.8mg/kg, about 1.8-1.9mg/kg, about 1.9-2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2- 2.3mg/kg, about 2.3-2.4mg/kg, about 2.4-2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7- 2.8mg/kg, about 2.8-2.9mg/kg, about 2.9-3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2- 3.3mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7- 3.8mg/kg, about 3.8-3.9mg/kg, 3.9-4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0- 7.0mg/kg, about 7.0-8.0mg/kg, about 8.0-9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg) dosage It is applied to experimenter.
In certain embodiments, the compound (or its pharmaceutically acceptable salt) of formula (I), such as, the chemical combination of the present invention Any one in thing 12-24, with about 1-20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2- 1.3mg/kg, about 1.3-1.4mg/kg, about 1.4-1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7- 1.8mg/kg, about 1.8-1.9mg/kg, about 1.9-2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2- 2.3mg/kg, about 2.3-2.4mg/kg, about 2.4-2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7- 2.8mg/kg, about 2.8-2.9mg/kg, about 2.9-3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2- 3.3mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7- 3.8mg/kg, about 3.8-3.9mg/kg, 3.9-4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0- 7.0mg/kg, about 7.0-8.0mg/kg, about 8.0-9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg) dosage It is used in the preparation of the medicine being applied to experimenter.
In another embodiment, present invention also offers a kind of for experimenter virus infect therapeutic and/or Preventative-therapeutic peroral dosage form, it comprises and has purity equal to or more than 91%w/w (such as, miscellaneous less than or equal to 9% Matter) the compound (or its pharmaceutically acceptable salt) of formula (I), such as, any one or its pharmacy in compound 12-24 Upper acceptable salt, wherein, when described compound is applied to the mankind with the dosage of about 2mg/kg when, described peroral dosage form About 2000 to about 4000h ng/mL of described compound, such as, the AUC of about 2500 to about 3000h ng/mL are provided.
In another embodiment, present invention also offers a kind of for experimenter virus infect therapeutic and/or Preventative-therapeutic peroral dosage form, it comprises and has the purity (e.g., less than or equal to 9% being equal to or greater than about 91%w/w Impurity) the compound (or its pharmaceutically acceptable salt) of formula (I), such as, any one or its medicine in compound 12-24 Acceptable salt on, wherein, when being executed with the dosage of about 1-2mg/kg, about 2-3mg/kg, about 3-4mg/kg by described compound With the when of to the mankind, described peroral dosage form provides the about 100-500h ng/mL of described compound, such as, about 200- The C of 400h ng/mLmax
In another embodiment, present invention also offers a kind of for experimenter virus infect therapeutic and/or Preventative-therapeutic peroral dosage form, it comprises and has the purity (e.g., less than or equal to 9% being equal to or greater than about 91%w/w Impurity) the compound (or its pharmaceutically acceptable salt) of formula (I), such as, any one or its medicine in compound 12-24 Acceptable salt on, wherein, when by described compound with the dosage of about 1-2mg/kg, about 2-3mg/kg, about 3-4mg/kg and The when that described compound being administered to the mankind to the metabolite of cidofovir, described peroral dosage form provides described cidofovir CmaxC less than described compoundmaxAbout 30%, such as, less than the C of described compoundmaxAbout 20%.
In certain embodiments, lasting ten accumulated doses are used.Such as, the compound of formula (I) is every with the dosage of about 100mg Week continue five weeks (that is, ten accumulated doses) for twice to be applied.Or, the compound of formula (I) can be with the loading dose of about 200mg The most about 100mg dosage is applied the most twice a week.In certain embodiments, lasting ten accumulated doses are used.Such as, formula (I) compound can be with the most about 9 other 100mg dosage the most totally ten the total agent of the loading dose of about 200mg Amount is applied.In one embodiment of the invention, the compound of formula (I) can press the scope of about 20-200mg/ days every day Dosage or weekly by the dosed administration of scope of about 200mg-2000mg.
When the compound of the present invention is administered to mammal (such as, the mankind) as medicine, they can give this Body or comprise as one, such as, about 0.1%-0.99%, about 0.2%-0.98%, about 0.3%-97%, about 0.4%- The pharmaceutical composition carrier pharmaceutically acceptable with one of the active component of 96% or about 0.5%-95% is combined.A reality Executing in example, the pharmaceutical composition of the active component comprising about 0.5%-95% carrier pharmaceutically acceptable with one is combined and is suitable for In being administered to mammal (such as, the mankind).Some embodiments of the present invention provide for treating, prevent or pre-anti-virus sense The preparation of the pharmaceutical composition of dye or the disease relevant with virus infection, described pharmaceutical composition comprise about 0.1%-99.9%, The compound of the formula (I) of about 0.2%-98%, about 0.3%-97%, about 0.4%-96% or about 0.5%-95% or its pharmaceutically Acceptable salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24.The invention provides about The change of the formula (I) of 0.1%-99.9%, about 0.2%-98%, about 0.3%-97%, about 0.4% to 96% or about 0.5%-95% Compound or its pharmaceutically acceptable salt (in such as compound 12-24 any one or its pharmaceutically acceptable salt) are comprising The compound of effective dose for treating, prevent or the manufacture of medicine of prophylaxis of viral infections or the disease relevant with virus infection In application.
According to conventional pharmaceutical compounding technique, the compound of formula of the present invention (I) or its pharmaceutically acceptable salt, Such as, any one or its pharmaceutically acceptable salt in compound 12-24, can tie mutually with pharmaceutically acceptable carrier Close.Additionally, described carrier can take many forms, this form depending on using required dosage form, such as, oral, per nasal, Rectum, vagina, parenteral (including intravenous injection or infusion).Preparation compositions for oral dosage form in any commonly employed Drug media can be used.Conventional drug media includes, such as, and water, glycol, oil, alcohol, flavoring agent, preservative, coloring Agent etc. are in the case of oral liquid (such as, suspension, solution, emulsion and elixir);Aerosol;Or carrier such as starch, Sugar, microcrystalline Cellulose, diluent, (the example in the case of oral solid formulation such as granulating agent, lubricant, binding agent, disintegrating agent As, powder, capsule and tablet).
The pharmaceutical composition of the compound (such as, the compound of formula (I)) comprising the present invention can be formulated into have appoints The concentration what is required.In certain embodiments, described compositions is formulated such that it comprises at least therapeutically effective amount.The present invention Described " therapeutically effective amount " instigates patient to improve necessary amount in clinical observation.In certain embodiments, described compositions It is formulated such that it to comprise not to cause the amount of one or more less desirable side effect.
Pharmaceutical composition includes being suitable to being administered orally, Sublingual, nose, rectum, vagina, locally, oral cavity and parenteral (include subcutaneous, Intramuscular and intravenous) use those, although most suitable approach will depend upon which character and the order of severity of the state of an illness for the treatment of.Institute State compositions can exist the most easily and be prepared by the method known to any pharmaceutical field.Real at some Executing in example, in order to Orally administered, described pharmaceutical composition is formulated into the form of pill, capsule, lozenge or tablet.Real at other Executing in example, described pharmaceutical composition is the form of suspension.
What dosage regimen can affect and constitute pharmacy effective dose.The compound of formula (I) or its pharmaceutically acceptable salt, example As, any one or its pharmaceutically acceptable salt in compound 12-24, can be administered to before or after seizure of disease Experimenter.It addition, several divided doses, and staggered dosage can every day or sequential application, or dosage can be continuously injected into, or It can be bolus injection.Add deduct additionally, described dosage proportionally can increase according to the urgency for the treatment of or the situation of prevention Few.And, described dosage can be with other antiviral drugs, such as, and well known to a person skilled in the art chemotherapeutant phase In conjunction with jointly using.Described reagent includes but not limited to, cloth woods cidofovir (BCV), ganciclovir (GCV), valganciclovir (vGCV), Le Taimowei (letermovir) and FOSCARNET and combinations thereof.Such as, the compound of formula (I) can with BCV, GCV, VGCV, Le Taimowei or FOSCARNET or combinations thereof combine and make for treating cmv infection and/or the disease relevant with CMV Or disease.In another embodiment, the compound of formula (I) can be used in combination with BCV and treat BKV (BK virus) infection And/or the disease relevant with BKV or disease.
The pharmaceutical composition of the present invention be formulated into its expected from route of administration match.The example of route of administration includes intestinal Outside stomach, such as intravenous, Intradermal, subcutaneous, oral (such as, sucking), percutaneous (locally) and mucosal administration.For parenteral, Intradermal or the solution of subcutaneous application or suspension can include following components: sterile diluent such as water for injection, saline solution, fixing Oil, Polyethylene Glycol, glycerol, propylene glycol or other synthetic;Antibacterial such as benzylalcohol or methyl parahydroxybenzoate;Antioxidant Such as ascorbic acid or sodium sulfite;Chelating agen such as ethylenediaminetetraacetic acid;Buffer agent such as acetate, citrate or phosphate, With tension regulator such as sodium chloride or dextrose.PH can be with acid or alkali, example hydrochloric acid or sodium hydroxide regulation.Parenteral administration Ampoule, disposable syringe or glass can be encapsulated in or in multiple dose vials that plastics are made.
At a preferred aspect, disease to be treated or situation are that virus infects.
For any compound, first therapeutically effective amount can cultivate detection, such as, tumor cell at cell, or dynamic Object model, it is common that rat, mice, rabbit, Canis familiaris L. or pig, middle estimation.Animal model can also be used to determine suitable concentration range And route of administration.Then such information may be used to determine whether effective dose and the approach used in the mankind.Treatment/prevention Curative effect and toxicity can be by cultivating or laboratory animal at cell, such as, and ED50(the effective agent for the treatment of in the 50% of colony Amount) and LD50The standard pharmaceutical procedures of (the lethal dosage in the 50% of colony) determines.Between toxicity and therapeutic effect Dose ratio is therapeutic index, and it can be expressed as ratio, LD50/ED50.The pharmaceutical composition showing big therapeutic index is Preferably.This dosage can change in this range according to the dosage form used, the sensitivity of patient and route of administration.
Regulate dosage and use enough levels of activating agent are provided or maintain intended effect.It is contemplated that factor include The seriousness of morbid state, the general health of experimenter, age, body weight and the sex of experimenter, diet, when using Between and frequency of administration, drug regimen (or multiple drug regimen), reaction sensibility and to treatment patience/reaction.Depot drug product Compositions according to the clearance rate of half-life and particular formulations can often 3-4 days, weekly, biweekly or be monthly applied.
In certain embodiments, use described in and continue ten accumulated doses.Such as, the compound of formula (I) can be with about 100mg Dosage twice a week continue five weeks (that is, ten accumulated doses) be applied.Or, the compound of formula (I) can be with about 200mg's Loading dose the most about 100mg dosage is the most double to be used.In certain embodiments, lasting ten accumulated doses are used.Example As, the compound of formula (I) can be with the loading dose of about 200mg then 9 other about 100mg dosage the most totally ten Accumulated dose is applied.In one embodiment of the invention, the compound of formula (I) can press the model of about 20-200mg/ days every day The dosage that encloses or weekly by the dosed administration of the scope of about 200mg-2000mg.
The pharmaceutical composition of the compound comprising the formula (I) of the present invention can in a generally known manner, such as, by often The mixing of rule, dissolve, pelletize, ingot processed, grinding, emulsifying, encapsulate, embed or prepared by freeze drying process.Pharmaceutical composition can be with Usual manner uses one or more include excipient and/or promote that reactive compound is processed into the pharmacy of the auxiliary agent of pharmaceutical formulation Upper acceptable carrier is prepared.Suitable preparation depends on the route of administration selected.
The pharmaceutical composition being suitable to injection use includes aseptic aqueous solution (in water miscible situation) or dispersion liquid and is used for The sterile powder of the interim preparation of sterile injectable solution or dispersion.Using for intravenous, suitable carrier includes physiology Saline, bacteriostatic water, polyoxyethylene castor oil ELTM(BASF, Pa Xipani, N.J.) or phosphate buffered saline (PBS) (PBS).All In the case of, compositions must be aseptic and should fluid to the degree easily existed with injectivity.It is producing and is storing Under the conditions of must be stable, and the contamination that must be prevented from microorganism such as antibacterial and fungus preserves.Described carrier can To be solvent or to contain, such as water, ethanol, polyhydric alcohol (such as, glycerol, propylene glycol and liquid macrogol etc.), and properly The disperse medium of mixture.Suitable mobility can be kept, such as, by using coating such as lecithin, by maintaining Granular size required in the case of a dispersion and by using surfactant.The prevention of microbial action can be by various Antibacterial and antifungal, such as parabens, methaform, phenol, ascorbic acid, thimerosal etc. realize.? In the case of many, preferably include isotonic agent, such as sugar, polyhydric alcohol such as mannitol, sorbitol, sodium chloride in the composition.Injection The absorption that postpones of compositions can be by comprising the reagent that can postpone to absorb in the composition, such as, aluminum monostearate and gelatin Realize.
Sterile injectable solution can be by enumerating the desired amount of reactive compound in a suitable solvent with above-mentioned Composition in one or a combination thereof mixing prepare, if it is desired, filtration sterilization subsequently.Usually, dispersant will be by living Property compound is incorporated into the sterile carrier containing basic disperse medium and prepares from other composition needed for above-mentioned enumerating.? In the case of preparing aseptic injectable solution with sterilized powder, preparation method is vacuum drying and lyophilization, and it produces activity and becomes The powder divided, adds from the most required any composition in the solution of its previous aseptic filtration.
Orally administered composition generally includes inert diluent or edible pharmaceutically acceptable carrier.They can be sealed It is contained in gelatine capsule or is pressed into tablet.In order to oral medication is used, reactive compound can mix with excipient and with sheet The form of agent, lozenge or capsule uses.Orally administered composition can also use the liquid-carrier as collutory to prepare, wherein, Compound in liquid-carrier is Orally administered, and wash, spue or swallow.The binding agent of pharmaceutically compatible and/or promoter material A part for compositions can be included as.Described tablet, pill, capsule, lozenge etc. can contain any following component or class Compound like character: binding agent such as microcrystalline Cellulose, gum tragacanth or gelatin;Excipient such as starch or lactose, disintegrating agent is such as Alginic acid, starch or corn starch;Lubricant such as magnesium stearate or Sterotes;Fluidizer such as silica sol;Sweeting agent is such as Sucrose or saccharin;Or flavoring agent, e.g., such as Herba Menthae, methyl salicylate or orange flavoring agent.
For being used by suction, described compound is with always self-pressurization container or allotter or the aerosol spray of aerosol apparatus The form transmission of mist, it comprises suitable propellant, such as, gas, such as carbon dioxide.
Systemic administration can also pass through through mucous membrane or transcutaneous modalities.For through mucous membrane or applied dermally, it is suitable as infiltration The penetrating agent on barrier layer is used in preparation.Such penetrating agent is the most generally known, and includes, such as, For mucosal administration, detergent, bile salts and fusidic acid derivatives.Mucosal administration can by use nasal spray or Suppository realizes.For applied dermally, reactive compound is configured to ointment as known in the art, ointment, gel Agent or emulsifiable paste.
Described compound can avoid, with protection compound, the pharmaceutically acceptable carrier that quickly runs off from health, such as Controlled release preparation, prepares including implant and microencapsulation transmission system.Polymerization biodegradable, biocompatible can be used Thing, such as ethylene vinyl acetate, polyanhydride, polyglycolic acid, collagen, poe and polylactic acid.For preparing the side of this type of preparation Method it will be apparent to those skilled in the art that.This material can also be buied from Alza company and Nova drugmaker.Lipid Liquid suspension (including the cell monoclonal antibodies liposome infected for the targeting of virus antigen) is also used as pharmaceutically may be used The carrier accepted.
The oral or parenteral compositions being configured to be prone to use dosage unit form uniform with dosage is advantageous particularly 's.Described dosage unit form refers to the discrete unit physically being suitable as single dose for treating experimenter;Each list Unit is containing being computed producing reactive compound predetermined of desired therapeutic effect to produce to combine with required pharmaceutical carrier Amount.For the specification of dosage unit form of the present invention by determining and directly depending on the specific characteristic of reactive compound and want real Existing specific therapeutic effect.
In treatment use, in other factors of the selected dosage of impact, pharmaceutical composition used according to the present invention Dosage depends on the clinical condition of medicament, age, body weight and receptor patient, and clinicist or the experience of doctor's administering therapeutic And judgment.Dosage can be from about 0.01mg/kg to about 100mg/kg.At preferred aspect, dosage can be from about 0.01mg/kg to about 10mg/kg.In one aspect, described dosage can be from about 1mg to about 1g;About 10mg to about 500mg; About 20mg to about 400mg;About 40mg to about 400mg;Or about 50mg to about 400mg, single, separate or continuous print dosage (its dosage can according to patient with the body weight of kg, with m2Body surface area and in units of year age regulation).Real at some Executing in example, the amount of every dosage form can be about 0.1mg to about 1000mg, such as, about 0.1mg, about 0.5mg, about 1mg, about 2mg, about 3mg, about 4mg, about 5mg, about 6mg, about 7mg, about 8mg, about 9mg, about 10mg, about 15mg, about 20mg, about 25mg, about 30mg, about 35mg, about 40mg, about 45mg, about 50mg, about 55mg, about 60mg, about 65mg, about 70mg, about 75mg, about 80mg, about 85mg, about 90mg, about 95mg, about 100mg, about 200mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 800mg, About 900mg or about 1000mg.In one embodiment, described amount can be about 20mg.In one embodiment, described amount is permissible It is about 50mg.In another embodiment, described dosage can be 100mg.
The compound of formula (I) or its pharmaceutically acceptable salt, such as, any one in the compound 12-24 of the present invention Kind or its pharmaceutically acceptable salt, be formulated into pharmaceutical composition or be used for preparation treatment virus infect and/or with virus Infect relevant disease and/or the medicine of disease.The compound of formula (I) or its pharmaceutically acceptable salt (such as compound 12- Any one or its pharmaceutically acceptable salt in 24) compositions and/or medicine be formulated into tablet or suspension.Formula (I) compound or its pharmaceutically acceptable salt (such as, in compound 12-24 any one or its pharmaceutically acceptable Salt) tablet be formulated into and comprise pharmacologically acceptable buffer agent, excipient, carrier, including emulsifying agent, reinforcing agent (such as, crospovidone (polyvinyl polypyrrolidone, PVPP, the friendship of (such as absorption enhancer), disintegrating agent Polyvidone, friendship polyvinyl pyrrolidone or E1202), it is the polyvinylpyrrolidone (PVP) of highly cross-linked modification), and/or at this Disclosed in bright and well-known polymer.
In one embodiment, the compound or its pharmaceutically acceptable salt that the invention provides formula (I) (such as, are changed Any one or its pharmaceutically acceptable salt in compound 12-24) tablet formulation in prophylactic treatment or prophylaxis of viral infections And/or the application in the disease relevant with virus infection or disease.The invention provides the compound of formula (I) or it pharmaceutically may be used The tablet formulation of the salt (such as, in compound 12-24 any one or its pharmaceutically acceptable salt) accepted needs in treatment The experimenter of this treatment (include but not limited to immunized subject, or the experimenter before or after organ and/or tissue transplantation) in Application.The invention provides the compound of formula (I) or its pharmaceutically acceptable salt (such as, any in compound 12-24 A kind of or its pharmaceutically acceptable salt) for treat needs the experimenter of this treatment (include but not limited to, immune tested Experimenter before or after person, or organ and/or tissue transplantation) medicine preparation in application.
In one embodiment, compound 12 or compound 13, or its pharmaceutically acceptable salt, it is formulated for pre- The treatment of anti-property or prophylaxis of viral infections and/or the disease relevant with virus infection or the tablet of disease.In certain embodiments, change Compound 12 or compound 13, or its pharmaceutically acceptable salt, be formulated for treat immunized subject, or organ and/or The tablet of the experimenter before or after tissue transplantation.In certain embodiments, compound 12 or compound 13, or it pharmaceutically can connect The salt being subject to, be formulated for prepare for treat need this treatment experimenter (include but not limited to immunized subject, or Experimenter before or after organ and/or tissue transplantation) the tablet of medicine.
In one embodiment, compound or its pharmaceutically acceptable salt of formula (I) (such as, is appointed in compound 12-24 Meaning is a kind of or its pharmaceutically acceptable salt) 100mg tablet formulation include silicified microcrystalline cellulose (Prosolv 90) (about 27.8%wt/ sheet), crospolyvinylpyrrolidone (Polyplasdone XL-10) (about 22%wt/ sheet), microcrystalline Cellulose and Mannitol (Avicel HFE102) (about 3.7%wt/ sheet), microcrystalline Cellulose (PHE102) (about 11.4%wt/ Sheet), mannitol (Pearlitol 100 SD) (about 33.9%wt/ sheet), silica sol(0.5% Wt/ sheet) and magnesium stearate (0.5%wt/ sheet).
In one embodiment, the compound or its pharmaceutically acceptable salt that the invention provides formula (I) (such as, are changed Any one or its pharmaceutically acceptable salt in compound 12-24) suspension preparation at prophylactic treatment or pre-anti-virus Infect and/or application in the disease relevant with virus infection and/or disease.The invention provides the compound of formula (I) or its The suspension preparation of pharmaceutically acceptable salt (such as, in compound 12-24 any one or its pharmaceutically acceptable salt) The experimenter needing this treatment in treatment (includes but not limited to immunized subject, or before or after organ and/or tissue transplantation Experimenter) in application.
In another embodiment, other excipient includes but not limited to sodium phosphate, diacidic base, citric acid (monohydrate) (about 0.06%wt), sodium citrate (about 0.10%wt), xanthan gum (about 0.04%wt), P-hydroxybenzoic acid first Ester (sodium salt) (about 0.17%wt), propyl p-hydroxybenzoate (sodium salt) (about 0.02%wt), sucralose (about 0.05%wt), Microcrystalline Cellulose and sodium carboxymethyl cellulose (VivaPur MCG591) (about 1.56%wt), high-fructose corn syrup (about 55% Wt), sour lime spice (WONF220J15) (about 0.40%wt), sodium hydroxide pellets, sodium hydroxide/hydrochloric acid and pure water are (about 68.93%wt).
The preparation of the present invention for the treatment End organ damage relevant with viral infection, such as, treat, prevent and/or Improve BKV relevant with End organ damage in experimenter to infect.
The preparation of the present invention is for preparing prophylactic treatment and/or prophylaxis of viral infections and/or the disease relevant with virus And/or the medicine of disease.
In one embodiment, compound or its pharmaceutically acceptable salt of formula (I) (such as, is appointed in compound 12-24 Meaning one or its pharmaceutically acceptable salt) it is applied twice a week with the dosage of about 100mg (tablet or suspension preparation).? In some embodiments, use and continue ten accumulated doses.Such as, the compound of formula (I) can with the dosage of about 100mg biweekly Continue five weeks (that is, ten accumulated doses) to be applied.Or, the compound of formula (I) can with the loading dose of about 200mg the most about 100mg dosage is the most double to be used.In certain embodiments, lasting ten accumulated doses are used.Such as, the chemical combination of formula (I) Thing can be applied with the loading dose of about 200mg then 9 other about 100mg dosage ten accumulated doses the most totally. In one embodiment of the invention, the compound of formula (I) can every day by the dosage or weekly of the scope of about 20-200mg/ days Dosed administration by the scope of about 200mg-2000mg.
In another embodiment, the compound of formula (I) or its pharmaceutically acceptable salt are (such as, in compound 12-24 Any one or its pharmaceutically acceptable salt) tablet or suspension, with dosage every day of about 40-1000mg, weekly (QW) or biweekly (BIW) is applied.In another embodiment, the compound of formula (I) or its pharmaceutically acceptable salt The tablet of (such as, in compound 12-24 any one or its pharmaceutically acceptable salt) or suspension, with about 40-400mg's Dosage every day, weekly (QW) or biweekly (BIW) are applied.
In another embodiment, the invention provides there is the compositions (example of gratifying pharmacokinetic properties As, pharmaceutical composition).Such as, the compositions of the present invention can provide the compound of formula (I) of blood level or its pharmaceutically can connect The salt that is subject to (such as, in compound 12-24 any one or its pharmaceutically acceptable salt), it is metabolized to therapeutic activity form After (such as, triphosphate equivalent), the blood level of the metabolite of generation will not inducing toxic (such as, nephrotoxicity).
The effective dose of medicament is to provide the objective identification that can be noticed by clinician or other qualified observers The amount improved.Term used herein " dosage effective means " refers to produce the activity of required biological effect in experimenter or cell The amount of compound.
In another embodiment, the compound of formula (I) or its pharmaceutically acceptable salt, such as, in compound 12-24 Any one or its pharmaceutically acceptable salt, is administered to experimenter with single dose.In another embodiment, formula (I) Compound or its pharmaceutically acceptable salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24, It is administered to experimenter with multiple dose.Multiple dose can periodically be used, and such as, every 12 hours are once, once a day, and every 2 days, often 3 days, every 4 days, every 5 days, every 6 days, every 7 days, every 8 days, every 9 days, every 10 days, every 11 days, every 12 days, every 13 days, every 14 days or Every 15 days.Such as, dosage can be applied twice a week.Additionally, each single dosage can be with same or different dosage It is applied.
Such as, any one or its pharmaceutically acceptable salt (example during experimenter can be applied the compound of formula (I) As, any one or its pharmaceutically acceptable salt in compound 12-24), first use about 1-20mg/kg (such as, about 1- 1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4-1.5mg/kg, about 1.5- 1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9-2.0mg/kg, about 2.0- 2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4-2.5mg/kg, about 2.5- 2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9-3.0mg/kg, about 3.0- 3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5- First dose of 3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9-4.0mg/kg Any one (or its pharmaceutically acceptable salt) in the compound 12-24 of amount, uses 1-4mg/ in this week or next week subsequently Kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4- 1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9- 2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4- 2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9- 3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5- 3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9-4.0mg/kg, about 4.0- 5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0-9.0mg/kg, about 9.0- Any one (or its medicine in 10.0mg/kg, or the compound 12-24 of the one or more other dosage of about 10-20mg/kg Acceptable salt on).Such as, experimenter can be applied first dosage of about 3mg/kg, uses the one of about 1mg/kg subsequently Individual or multiple other dosage.Such as, experimenter can be applied first dosage of about 2mg/kg, uses the one of about 3mg/kg subsequently Individual or multiple other dosage.Such as, experimenter can be applied first dosage of about 4mg/kg, the about 4mg/kg's used subsequently One or more other dosage.
Multiple dose can also be used at variable time interval.Such as, the 2nd, 3,4,5,6,7 or 8 or more dosage can To be applied with 6 days intervals, it is applied other dosage with 7 days intervals subsequently.Such as, the 2nd, 3,4,5,6,7 or 8 or more Individual dosage can be applied with 7 days intervals, is applied other dosage with 3 days intervals subsequently.
In one embodiment, the compound of formula (I) or its pharmaceutically acceptable salt, such as, compound 12-24 appoints Meaning one or its pharmaceutically acceptable salt, the dosage with about 40-1000mg is weekly, or the dosage with about 40-1000mg It is administered to experimenter twice a week.
In a further embodiment, the invention provides a kind of delaying to fall ill, reduce risk or the experimenter infecting BKV Middle treatment end organ damage or the method for damage, described method includes to the Orally administered choosing comprising therapeutically effective amount of experimenter Any one compound in compound 12-24 or its pharmaceutically acceptable salt.
In certain embodiments, it is experimenter after HSCT with the experimenter of one or more compounds for treating of the present invention. In other embodiments, with the experimenter of one or more compounds for treating of the present invention, there is End organ damage, wherein, be subject to The organ of impact includes but not limited to kidney, ureter, bladder, prostate or urethra.
In certain embodiments, it is HCV experimenter with the experimenter of one or more compounds for treating of the present invention.At it In his embodiment, with the experimenter of one or more compounds for treating of the present invention, there is End organ damage, wherein, impacted Organ include but not limited to kidney, ureter, bladder, prostate or urethra.
In certain embodiments, the invention provides a kind of by the Orally administered choosing including therapeutically effective amount of experimenter Any one compound in compound 12-24 or its pharmaceutically acceptable salt reduce the method for the sickness rate of HCV.
In certain embodiments, the invention provides a kind of by exist BKV infect reactivation risk experimenter's mouth Clothes are used and are included any one compound in compound 12-24 of therapeutically effective amount or its pharmaceutically acceptable salt Pharmaceutical composition reduces the method for the sickness rate of hematuria or renal function injury.
In certain embodiments, the compound of the present invention reduces in the experimenter that there is BKV infection reactivation risk Hematuria or the sickness rate of renal function injury, wherein said experimenter is experimenter after HSCT.In a further embodiment, this The bright BKV that provides for reducing in described experimenter infects the compound of the formula (I) of reactivation or it is pharmaceutically acceptable The pharmaceutical composition of salt.The pharmaceutical composition of the present invention reduces the BK virus carrying capacity in experimenter, and delays morbidity or reduce End organ damage or the risk of infringement.End-organ includes but not limited to kidney, ureter, bladder, prostate and urethra.
In certain embodiments, the pharmaceutical composition every day of the present invention, weekly (QW) or twice a week (BIW) with about The compound (such as, compound 12-24 or its pharmaceutically acceptable salt) of the formula (I) of 40-1000mg is applied.The present invention's Pharmaceutical composition every day, weekly (QW) or twice a week (BIW) with about 40mg, 50mg, 75mg, 100mg, 150mg, 175mg、200mg、250mg、275mg、300mg、325mg、350mg、375mg、400mg、450mg、500mg、500-600mg、 600-700mg, 700-800mg, 800-900mg or 900-1000mg, or twice a week with about 40mg, 50mg, 75mg, 100mg, 150mg, 175mg, 200mg, 250mg, 275mg, 300mg, 325mg, 350mg, 375mg or 400mg, 450mg, 500mg, 500- The compound 12-24 of 600mg, 600-700mg, 700-800mg, 800-900mg or 900-1000mg or it is pharmaceutically acceptable Salt, be applied.
The compound of the present invention after HSCT after the 1st, 2,3,4,5,6,7 or to 10 days with 1-20mg/kg, such as, The dosage of 1.25mg/kg, 2.5mg/kg, 5.0mg/kg, 10.0mg/kg, or 20.0mg/kg is applied.This of 1-20mg/kg Bright compound can be applied once in a week or twice a week.In one embodiment, every with 1-20mg/kg of described treatment Week, applied once started, and then two weeks of 1-20mg/kg are used until meeting needs.
The invention provides with the dosage of about 1-20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4-1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9-2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4-2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9-3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9-4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0-9.0mg/kg, about 9.0-10.0mg/kg, about 10-15mg/kg, or about The compound of the formula (I) 15-20mg/kg) used, such as, in compound 12-24 any one (or it is pharmaceutically acceptable Salt).
In certain embodiments, the invention provides the compound of the formula (I) being formulated into pharmaceutical composition, such as, change In compound 12-24 any one (or pharmaceutically acceptable salt).In one embodiment, the compound of formula (I), such as, change In compound 12-24, any one (or pharmaceutically acceptable salt) is formulated into tablet.In another embodiment, formula (I) Compound, such as, in compound 12-24, any one (or pharmaceutically acceptable salt) is formulated into suspension.
The invention provides treatment and/or the prevention of the virus infection of the compound using the present invention.Change shown in formula (I) Compound is used for treatment, prevents at least one virus and/or prepare the medicine for treating and/or prevent at least one virus, At least one virus described is selected from adenovirus, CMV, JCV, BKV, SV40, MCV, KIV, WUV, EBV, vaccinia virus, herpes simplex Virus 1 type (HSV-1), herpes simplex virus type 2 (HSV-2), human herpes virus-6 (HHV-6), Human herpesvirus 8 (HHV-8), hepatitis B virus, hepatitis C virus, varicella zoster virus (VZV), classical smallpox, alastrim, variola, Cowpox, camel pox, monkeypox, poliovirus, Ebola virus, Marburg virus, enterovirus (such as, EV68 and EV71), papillomavirus, HIV (human immunodeficiency virus) (HIV), influenza and combinations thereof.
In certain embodiments, the invention provides a kind of passing through to the Orally administered therapeutically effective amount of experimenter in need Treat selected from the compound of compound 12-24 or the pharmaceutical composition of its pharmaceutically acceptable salt, prevent or delay CMV Infect or the disease relevant with cmv infection or the method for disease.
In certain embodiments, the invention provides a kind of passing through to the Orally administered therapeutically effective amount of experimenter in need Treat selected from the compound of compound 12-24 or the pharmaceutical composition of its pharmaceutically acceptable salt, prevent or delay HCV Infect or the disease relevant with HCV infection or the method for disease.
In certain embodiments, the invention provides a kind of passing through to the Orally administered therapeutically effective amount of experimenter in need Treat selected from the compound of compound 12-24 or the pharmaceutical composition of its pharmaceutically acceptable salt, prevent or delay horse Your fort virus infects or the disease relevant with Marburg virus infection or the method for disease.
In certain embodiments, the invention provides a kind of passing through to the Orally administered therapeutically effective amount of experimenter in need Treat, prevent or delay selected from the compound of compound 12-24 or the pharmaceutical composition of its pharmaceutically acceptable salt angstrom Win and draw virus infection or the disease relevant with Ebola virus infection or the method for disease.
In certain embodiments, the invention provides a kind of passing through to the Orally administered therapeutically effective amount of experimenter in need Treat selected from the compound of compound 12-24 or the pharmaceutical composition of its pharmaceutically acceptable salt, prevent or delay intestinal Road virus infects or the disease relevant with enterovirus infection or the method for disease.
Treatment virus infects (such as, cmv infection or the disease relevant with cmv infection or disease or HCV infection or and HCV Infect relevant disease or disease) experimenter weekly or be administered twice weekly about 40mg, 50mg, 75mg, 100mg, The compound selected from compound 12-24 of 150mg, 175mg, 200mg or 250mg or its pharmaceutically acceptable salt.The present invention Provide cmv infection or the disease relevant with cmv infection or disease or HCV infection or the disease relevant with HCV infection or disease The treatment of the experimenter of disease, by using (QW) about 200mg once in a week or (BIW) about 100mg twice a week to experimenter Compound or its pharmaceutically acceptable salt selected from compound 12-24.In one embodiment, twice a week (BIW) is with about The compounds for treating experimenter of 100mg.In another embodiment, weekly (QW) with about 200mg or twice a week (BIW) With the compounds for treating experimenter of about 100mg.Cmv infection or the disease relevant with cmv infection or disease or HCV infection or with Disease or the experimenter of the treatment of disease that HCV infection is relevant are HSCT experimenters and accept allogeneic stem cell transplantation.
Present invention also offers a kind of by using the change selected from compound 12-24 including therapeutically effective amount to experimenter The pharmaceutical composition of compound or its pharmaceutically acceptable salt carrys out prophylactic treatment, prevents or delay cmv infection or and cmv infection Relevant disease or the method for disease.
Present invention also offers a kind of by using the change selected from compound 12-24 including therapeutically effective amount to experimenter The pharmaceutical composition of compound or its pharmaceutically acceptable salt carrys out prophylactic treatment, prevents or delay HCV infection or and HCV infection Relevant disease or the method for disease.
Invention further provides a kind of prophylactic treatment, prevent or delay Marburg virus to infect or and Marburg Poison infects relevant disease or the method for disease, by use to experimenter include therapeutically effective amount selected from compound 12-24 Compound or the pharmaceutical composition of its pharmaceutically acceptable salt, and combine and use selected from immunosuppressant and antiviral agent One or more compounds or compositions.
Invention further provides a kind of prophylactic treatment, prevention, or delay Ebola virus to infect or and Ebola Virus infects relevant disease or the method for disease, by use to experimenter include therapeutically effective amount selected from compound 12- A kind of compound in 24 or the pharmaceutical composition of its pharmaceutically acceptable salt, and combine and use selected from immunosuppressant and anti- One or more compounds in viral agent or compositions.
Invention further provides a kind of prophylactic treatment, prevent or delay enterovirus infection or with enterovirus sense Be infected with the disease of pass or the method for disease, by use to experimenter include therapeutically effective amount in compound 12-24 A kind of compound or the pharmaceutical composition of its pharmaceutically acceptable salt, and combine use selected from immunosuppressant and antiviral agent One or more compounds or compositions.
Invention further provides a kind of prophylactic treatment, prevent or delay virus to infect or relevant with virus infection Disease or disease (such as, human cytomegalovirus (HCMV), BK virus (BKV), epstein-Barr virus (EBV), adenopathy Poison, JC virus (JCV), SV40, MC virus (MCV), KI virus (KIV), WU virus (WUV), vaccinia virus, herpes simplex virus 1 type (HSV-1), herpes simplex virus type 2 (HSV-2), human herpes virus-6 (HHV-6), human herpes virus type 8 (HHV- 8), hepatitis B virus, hepatitis C virus, varicella zoster virus (VZV), classical smallpox, alastrim, variola, cowpox, Camel pox, monkeypox, poliovirus, Ebola virus, Marburg virus, enterovirus, papillomavirus and people Para-immunity defective virus (HIV)) method, by experimenter Orally administered include therapeutically effective amount selected from compound 12- The compound of 24 or the pharmaceutical composition of its pharmaceutically acceptable salt, and combine use selected from immunosuppressant and antiviral agent One or more compounds or compositions.
In certain embodiments, the pharmaceutical composition of the present invention with selected from midazolam, Ciclosporin A, tacrolimus, more VCV, valganciclovir, foscarnet sodium, cidofovir, two wires anti-CMV medicine, two wires anti-HCV medicament, FOSCARNET, Fei Gesi Booth, Pei Feisi booth, glucocorticoid such as budesonide, beclometasone, wide spectrum CYP inhibitor amino benzotriazole or a combination thereof One or more compounds or compositions combine to be used.
In certain embodiments, the invention still further relates to use the compound (such as, compound 12-24) of a kind of present invention or Such as JCV relevant for the PV of its pharmaceutically acceptable salt relevant, multifocal leukoencephalopathy (PVML) or the kidney relevant to PV Sick treatment.
The invention provides compound or its pharmaceutically acceptable salt treatment experimenter of use formula (I), wherein, tested Person is immunocompromised host.In one embodiment, the experimenter of described immunocompromised host is the transplant patient of immunosuppressive drug.? In some embodiments, experimenter's infected by HIV of immunocompromised host.
In a further embodiment, other immunosuppressant of compound and at least one is combined and uses.An enforcement In example, immunosuppressant is administered simultaneously or sequentially.Immunosuppressant include but not limited to daclizumab, basiliximab, he Ke Mosi, sirolimus, mycophenolate, cyclosporin A, glucocorticoid, CD 3-resisting monoclonal antibody, anti-thymocyte ball egg In vain, anti-CD52 monoclonal antibody, azathioprine, everolimus, dactinomycin, cyclophosphamide, platinum phosphamide, nitroso ureas, first Aminopterin, mercaptopurine, not Luo Dankang, interferon gamma, infliximab, Embrel, adalimumab, his pearl monoclonal antibody, sweet smell Ge Mode and combinations thereof.
In another embodiment, the invention provides a kind of therapeutic that virus infects in experimenter and/or pre- The peroral dosage form of anti-property treatment, it comprises purity and is equal to or greater than about the compound of formula (I) of 91% or it is pharmaceutically acceptable Salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24, wherein, described peroral dosage form, with about 1- 20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4- 1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9- 2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4- 2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9- 3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4- 3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9- 4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0- 9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg)) the described compound of dosage be administered to the mankind after, it is provided that The EC of about 0.06-0.04 for the resistant mutants AD169 of HCMV UL5450(μM), such as, about 0.059,0.058, 0.057,0.056,0.055,0.054,0.053,0.052,0.051,0.050,0.049,0.048,0.047,0.046, 0.045,0.044,0.043,0.042,0.041 or 0.040.In one embodiment, for the resistant mutants of HCMV UL54 The compound 12 of AD169 and/or the EC of compound 1350(μM) is about 0.052.EC50By any method as known in the art Determine, and as described herein in the examples.
In another embodiment, the invention provides a kind of therapeutic that virus infects in experimenter and/or pre- The peroral dosage form of anti-property treatment, it comprises purity and is equal to or greater than about the compound of formula (I) of 91% or it is pharmaceutically acceptable Salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24, wherein, described peroral dosage form, with about 1- 20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4- 1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9- 2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4- 2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9- 3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4- 3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9- 4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0- 9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg)) the described compound of dosage be administered to the mankind after, it is provided that The EC of about 6.0-4.0 for the resistant mutants D542E of HCMV UL5450μM, such as, about 6.0-5.9,5.9-5.8,5.8- 5.7、5.7-5.6、5.6-5.5、5.5-5.4、5.4-5.3、5.3-5.2、5.2-5.1、5.1-5.0、5.0-4.9、4.9-4.8、 4.8-4.7,4.7-4.6,4.6-4.5,4.5-4.4,4.4-4.3,4.3-4.2,4.2-4.1 or 4.1-4.0.An embodiment In, for the compound 12 of resistant mutants D542E and/or the EC of compound 13 of HCMV UL5450(μM) is about 5.41.
In another embodiment, the invention provides a kind of mouth in experimenter virus infect therapeutic and/or Preventative-therapeutic oral dosage form, it comprises purity and is equal to or greater than about the compound of formula (I) of 91% or it is pharmaceutically acceptable Salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24, wherein, described peroral dosage form, with about 1- 20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4- 1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9- 2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4- 2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9- 3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4- 3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9- 4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0- 9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg)) the described compound of dosage be administered to the mankind after, it is provided that Resistant mutants GDF for HCMV UL54RThe EC of the about 0.2-0.15 of P5350μM, such as, about 0.2-0.19,0.19- 0.18,0.18-0.17,0.17-0.16 or 0.16-0.15.In one embodiment, for the resistant mutants of HCMV UL54 GDFRThe compound 12 of P53 and/or the EC of compound 1350(μM) is about 0.171.
In another embodiment, the invention provides a kind of therapeutic that virus infects in experimenter and/or pre- The peroral dosage form of anti-property treatment, it comprises purity and is equal to or greater than about the compound of formula (I) of 91% or it is pharmaceutically acceptable Salt, such as, any one or its pharmaceutically acceptable salt in compound 12-24, wherein, described peroral dosage form, with about 1- 20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4- 1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9- 2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4- 2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9- 3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4- 3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9- 4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0- 9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/kg)) the described compound of dosage be administered to the mankind after, it is provided that Resistant mutants 4955 for HCMV UL54RThe EC of about 0.2-0.1150μM, such as, about 0.2-0.19,0.19-0.18, 0.18-0.17,0.17-0.16,0.16-0.15,0.15-0.14,0.14-0.13,0.13-0.12, or 0.12-0.11.One In individual embodiment, for the resistant mutants 4955 of HCMV UL54RCompound 12 and/or the EC of compound 1350(μM) is about 0.143。
In certain embodiments, the compound of the present invention has anti-various virus (such as, herpes simplex virus and HCMV) Activity.Biologic activity detection method can be carried out in many systems, and such as, human foreskin fibroblast or Testis et Pentis Canis pass on (MDCK) cell.More bioactive examples of the compound of the present invention are provided below.
Table 2 is compound 12 anti-herpes simplex virus 1 (E-377 strain) and HCMV (AD169 in human foreskin fibroblast Strain) activity
Table 3 is compound 12 and six decyloxy propyl group-cidofovir (HDP-CDV) anti-HCMV in human foreskin fibroblast The activity of the disease resistance mutant of UL54
Table 4 is compound 12 anti-vaccinia virus (Copenhagen strain) and vaccinia virus (Bu Lai in human foreskin fibroblast Strain) activity
Table 5 is the activity of compound 12 resisiting influenza virus strain in Testis et Pentis Canis passes on (MDCK) cell
Table 6 is the activity of the anti-epstein-Barr virus of compound 12 in A Kata cell (Akata cells)
Table 7 in MRC-5 cell anti-HCMV and in African green monkey kidney cell the compound 12 and HDP-CDV of anti-BKV Activity
The Anti-viral activity in vitro of table 8 compound 12
Table 9 Anti-viral activity in vitro compares (meansigma methods XC50)
The invention provides compound or its pharmaceutically acceptable salt (such as, appointing in compound 12-24 of formula (I) Anticipate a kind of compound or its pharmaceutically acceptable salt), it has the curative effect/toxicity ratio of improvement compared with HDP-CDV.One In a little embodiments, the compound of the present invention is to SI value between 900-100 and/or to BKV of selectivity index (SI) value of CMV Between 15-20.In one embodiment, SI (selectivity index) value of CMV is about by compound 12 and/or compound 13 948 and/or BKV is about 16.5, and HDP-CDV is about 150 to the SI value of CMV and BKV is about 3.3.
In another embodiment, the invention provides a kind of therapeutic that virus infects in experimenter and/or pre- The peroral dosage form of anti-property treatment, it comprises purity and is equal to or greater than about in the compound 12-24 of 91% any one or its pharmacy Upper acceptable salt, wherein, described peroral dosage form, with about 1-20mg/kg (such as, about 1-1.1mg/kg, about 1.1-1.2mg/ Kg, about 1.2-1.3mg/kg, about 1.3-1.4mg/kg, about 1.4-1.5mg/kg, about 1.5-1.6mg/kg, about 1.6-1.7mg/ Kg, about 1.7-1.8mg/kg, about 1.8-1.9mg/kg, about 1.9-2.0mg/kg, about 2.0-2.1mg/kg, about 2.1-2.2mg/ Kg, about 2.2-2.3mg/kg, about 2.3-2.4mg/kg, about 2.4-2.5mg/kg, about 2.5-2.6mg/kg, about 2.6-2.7mg/ Kg, about 2.7-2.8mg/kg, about 2.8-2.9mg/kg, about 2.9-3.0mg/kg, about 3.0-3.1mg/kg, about 3.1-3.2mg/ Kg, about 3.2-3.3mg/kg, about 3.3-3.4mg/kg, about 3.4-3.5mg/kg, about 3.5-3.6mg/kg, about 3.6-3.7mg/ Kg, about 3.7-3.8mg/kg, about 3.8-3.9mg/kg, about 3.9-4.0mg/kg, about 4.0-5.0mg/kg, about 5.0-6.0mg/ Kg, about 6.0-7.0mg/kg, about 7.0-8.0mg/kg, about 8.0-9.0mg/kg, about 9.0-10.0mg/kg, or about 10-20mg/ The compound 12-24 of dosage kg)) appoints in any one (or its pharmaceutically acceptable salt) and described compound 12-24 After the metabolite that a kind of (or its pharmaceutically acceptable salt) metabolism is triphosphate equivalent of anticipating is administered to the mankind, it is provided that few The C of any one described compound in compound 12-24 (or its pharmaceutically acceptable salt)maxAbout 30% described The C of triphosphate equivalentmax, such as, less than the C of described compoundmaxAbout 20%.In certain embodiments, described metabolite The C of (that is, triphosphate equivalent)maxLess than any one the C in compound 12-24maxAbout 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15% or 10%.
The pharmacokinetics of compositions can some change from experimenter to experimenter in crowd.Above-mentioned for this The average behavior that the bright numeral described in compositions is based in crowd.It is contemplated that include that meansigma methods falls in the present invention In the range of compositions, although it will be appreciated that some main body may fall outside scope.
Described pharmaceutical composition can be included in container, packaging or allotter together with operation instructions.The present invention provides A kind of include the container together with operation instructions except any one the pharmaceutical composition in disclosed compound, Packaging or the test kit of allotter.
The compound of the present invention can form salt further.All these forms is contemplated in claimed invention Within the scope of.
Prevent the disease due to virus reactivation or the method for disease
Present invention also offers a kind of prevention disease or side of disease in the experimenter having virus to infect reactivation danger Method, by compound or its pharmaceutically acceptable salt (example of the formula (I) to described experimenter Orally administered treatment effective dose Such as any one or its pharmaceutically acceptable salt in compound 12-24) pharmaceutical composition.In certain embodiments, have again Activating dangerous virus can be BKV.In some preferred embodiments, the virus having reactivation dangerous can be CMV.
In one embodiment, having virus to infect the dangerous experimenter of reactivation can be stem cell transplantation or nephrocyte shifting Plant receiver.In one embodiment, experimenter after experimenter can be HSCT.In other embodiments, experimenter can be Islet cell transplantation receiver, bone marrow transplantation receiver, endotheliocyte are transplanted receiver, epidermis cell transplanting receiver, are become flesh Stem cell receiver that cell transplantation receiver, muscle are derivative and/or neural stem cells transplantation receiver.
In other embodiments, experimenter can be islet cell transplantation receiver, bone marrow transplantation receiver, endotheliocyte Transplant receiver, epidermis cell transplants receiver, myoblast transplantation receiver and/or neural stem cells transplantation receiver.
In another embodiment, the inventive method prevents at HSCT by the hematuria in rear examination person or renal function injury.? After HSCT, in patient, hematuria is relevant with the prevention that the virus in experimenter infects with the prevention of renal function injury.An embodiment In, virus infects the prevention of reactivation and prevents hematuria and renal function injury in described experimenter.
The method reducing the sickness rate of the BKV relevant with hematuria and/or renal function injury
The method that the application further relates to reduce the sickness rate of the BKV relevant with hematuria and/or renal function injury.The present invention's Method prevents all relevant with the End organ damage infected from BKV hematuria and the appearance of renal function injury.The present invention is also Relate to the compound by formula (I) or its pharmaceutically acceptable salt (such as, compound 12 or compound 13, or it pharmaceutically may be used The salt accepted) reduce in patient, BK virus load increases after HSCT danger and/or delay HSCT after BK virus load in patient Increase, thus in these patients, reduce the danger of end-organ disease and/or the method delaying end-organ disease.The present invention Pharmaceutical composition can prevent end organ damage or damage, such as, kidney, ureter, bladder, prostate and urethra infringement or Damage.
In certain embodiments, the method for the sickness rate reducing the BKV relevant with hematuria and/or renal function injury provides By weekly for the compound of the present invention of about 40-100mg (QW) or twice a week (BIW) be applied to experimenter and prevent or control Treat end organ damage or damage.In one embodiment, with about 40-1000mg QW or BIW or about 100-200mg on every Mondays Secondary or two treatments experimenter.Infected experimenter's every day of dsDNA virus (such as, BKV), weekly (QW) uses about 40- 100mg or twice a week (BIW) with the compound of the present invention of about 40-1000mg (such as, compound 12 or compound 13, or Its pharmaceutically acceptable salt) treatment,.In a further embodiment, every day, weekly (QW) about 150mg or about 200mg, or twice a week (BIW) with compound (such as, compound 12 or the compound of the present invention of about 75mg or about 100mg 13, or its pharmaceutically acceptable salt) treat described experimenter.
In other embodiments, the method being used for reducing the sickness rate of the BKV relevant with hematuria and/or renal function injury carries For the dosage treatment experimenter of about 50-99mg, 101-149mg, 151-199mg, 201-250mg, or > 251mg, do not have Bring significant adverse effect (AEs).In certain embodiments, described dosage one week, within two weeks or in whole treatment week Interim change.
Sickness rate based on treatment burst hematuria have evaluated the compound of formula (I) or its pharmaceutically acceptable salt to hemorrhage The impact of the appearance of property cystitis.The invention provides and exist with compound 12 or compound 13 (or its pharmaceutically acceptable salt) The HSCT patient that baseline (BKU+) BKV is positive prevents hematuria (Hem+).In one embodiment, compared with placebo group, Hem+ in the patient of baseline BKV negative (BKU-) is not significantly different from.
Measure the compound of formula (I) or its pharmaceutically acceptable salt to previously existing in the experimenter that BKV infects The impact of renal function injury.Such as, the present invention provides pre-in the patient of baseline (after HSCT transplants) BKV viruria (BKU+) The increase of anti-creatinine levels and the deterioration of renal function, and compared with placebo group, described patient with compound 12 or 13 (or its Pharmaceutically acceptable salt) treatment.In one embodiment, compound 12 or compound 13 (or its pharmaceutically acceptable salt) Do not affect the end organ damage in the patient of baseline BKV viruria negative (BKU-).In these patients, with placebo group Comparing, described creatinine levels does not increases.
The invention provides the compound of use formula (I) or its pharmaceutically acceptable salt reduces and comes off in its urine BKV viruria experimenter in microscopic hematuria.Such as, during treating, i.e. accept compound 12 13 or its pharmaceutically The pharmaceutical composition of acceptable salt, suffers from the experimenter of BKV viruria, compares the experimenter accepting placebo, blood sun Property urine examination decline 2-10 times.In certain embodiments, between treatment group and placebo group, the difference of blood positive urine examination may It is 2-8 times, 2-7 times, 2-6 times, 2-5 times or 2-4 times.In the experimenter not having BK virus urine disease, the patient or not for the treatment of Between the patient (such as accepting the patient of placebo) for the treatment of, the ratio of blood positive urine examination is probably low or suitable.
The BK relevant with active urine bladder with the compound of formula (I) or the method pair of its pharmaceutically acceptable salt treatment has Beneficial effect.Such as, high BK virus urine disease measures (such as, >=1 × 1010Copy/mL) with the most important activity (example As, AEs is band blood in cystitis or urine) relevant.Compare the experimenter of placebo treatment, the blood positive urinalysis of confirmation Ratio can occur at compound 12 or the 1/ of the ratio of the experimenter of the treatment of compound 13 (or its pharmaceutically acceptable salt) 10、1/9、1/8、1/7、1/6、1/5、1/4.In certain embodiments, for compound 12 or compound 13, (or it is pharmaceutically Acceptable salt) experimenter developing BK virus urine disease during treating that treats, the sickness rate of lasting BK virus urine disease can It is reduced.
The method of the embodiment of the present invention relates to the measurement of the concentration of the serum creatinine as renal function mark.The present invention Method weighs renal function by kidney from internal creatinine clearance by calculating.This is referred to as creatinine clearance, and it Estimate the speed filtered by kidney (glomerular filtration, or GFR).Creatinine clearance rate is measured in two ways.It is by using Serum (blood) creatinine values, the body weight of patient and the formula at age calculate.Creatinine clearance is also by collecting 24 hours Urine specimen measures.In blood, the normal level of kreatinin for 0.7-1.3mg/dL and is 0.6-for women for male 1.1mg/dL.See Creatinine Blood, Medline Plus, U.S.National Library of Medicine, NIH.If renal function is abnormal, the creatinine levels in blood will increase (because almost without the kreatinin urine by you Release).
Creatinine levels in urine exceedes about 1.36mg/mL and is considered to raise.In the method for the invention, in the treatment phase Between creatinine levels be considered as the most important change from the increase of baseline about 15% or about 25%.
The inventive process provides the evaluation of use haemachrome+1 urine examination microscopic hematuria as an alternative.At serum creatinine In measurement treatment terminate (last value) improve (such as, > 120 μMs (1.36mg/dl)) be considered to have clinical meaning.Logical Crossing the last value measuring kreatinin, it is higher than normal value, such as, more than > 120 μMs with from baseline at least 15% or the increasing of 25% Add, distinguish the renal dysfunction being pre-existing in.
The inventive process provides by Orally administered compound 12 or compound 13 that (or it is pharmaceutically acceptable Salt) reduce the danger of end organ damage in BKV positive patient or delay the end organ damage in BKV positive patient Morbidity.It is that the experimenter of BV viruria is owing to compound 12 or compound 13, (or it pharmaceutically can connect during treating The salt being subject to) treatment can show beneficial effect, reduce the morbidity of the renal dysfunction (kreatinin rising) of 1.5-4.5 times Rate.The sickness rate of renal dysfunction can be reduced about 1.6,1.7,1.8,1.9,2.0,2.1,2.2,2.3,2.4,2.5,2.6, 2.7,2.8,2.9,3.0,3.1,3.2,3.3,3.4,3.5,3.6,3.7,3.8,3.9,4.0,4.1,4.2,4.3,4.4 or 4.5 Times.In the BK positive subjects of the present invention, it is understood that there may be kreatinin raises or newly send out 1.2-4.4 times of haemachrome+urine examination Reduce.Keep in negative for BK experimenter during treating, kreatinin is raised or kreatinin or hemoglobinuria+total score The ratio of analysis may be the most similar.
Renal function and bladder are had an impact (hematuria, cystitis, dysuria etc.) by BKV.Analysis (the serum of routine test value Kreatinin raises and the new existence sent out, it was demonstrated that hematuria) experimenter provides after HSCT the potential mark of BK impact.This Bright method provides in the experimenter treated with compound 12 or compound 13 (its pharmaceutically acceptable salt) in treatment Period measures the end value of kreatinin, and creatinine levels exceedes increase % and haemachrome+1 urine examination of baseline.
Therapeutic alliance
Compound provided by the present invention or compositions can also be with reinforcing agents, and other active component, or press down with immunity Formulation compositions uses.In certain embodiments, compound can be with other therapeutic agent or reagents recombination, or sequential application. This type of other therapeutic agent includes for treating, prevent or improve known to one or more diseases relevant with virus infection Those.It should be appreciated that compound provided by the present invention and one or more compounds mentioned above and optional The combination of any appropriate of kind or other pharmacological active substances multiple is recognized as within the scope of the present invention.At another In individual embodiment, compound provided by the present invention was used before or after the active component that one or more are other.One In individual embodiment, two or more antiviral agent disclosed by the invention is continuously or combined administration.
The amount of some reinforcing agents can use method as known in the art to select to strengthen the biology of antiviral agent Availability.Any amount being provided that desired reaction by some reinforcing agents can be used.In nonrestrictive example, agent Amount can be the compound per kilogram body weight every day from 0.001mg to about 2000mg, such as, 0.01-500mg/kg, or such as 0.1-20mg/kg。
The compound used altogether with another kind of medicament or compositions that the present invention provides infect at treatment BKV, the swashing again of BKV Live or prevent that the end organ damage in the experimenter having infected BKV or damage have cooperative effect.Such combination Instantiation includes but not limited to: compound 12 or compound 13 (or its pharmaceutically acceptable salt) press down with at least one immunity Preparation combines.Exemplary immunosuppressant includes but not limited to, daclizumab, basiliximab, tacrolimus, Xi Luomo Department, mycophenolate (as wheat examines phenol sodium or mycophenolate), Ciclosporin A, glucocorticoid, CD 3-resisting monoclonal antibody (OKT3), Antithymocyte globulin (ATG), anti-CD52 monoclonal antibody (alemtuzumab 1-H), azathioprine, everolimus, more mildew Element, cyclophosphamide, platinum, nitroso ureas, methotrexate, azathioprine, purinethol, not Luo Dankang, interferon gamma, Infliximab Monoclonal antibody, Embrel, adalimumab, natalizumab (Tysabri or Natalizumab), FTY720 and combinations thereof.? In some embodiments, described pharmaceutical composition includes, such as, compound 12, natalizumab (Tysabri or And pharmaceutically acceptable carrier Natalizumab).
In one embodiment, pharmaceutical composition of the present invention includes, such as, compound 12 or compound 13 (or Its pharmaceutically acceptable salt) and one or more be used for treating virus infection, such as, cause progressive multifocal leukoencephalopathy The medicine of polyoma virus JC virus (" JCV ") of (" PML "), at least one pharmaceutically acceptable carrier.A reality Executing in example, one or more medicines described are selected from comprising(efalizumab),(his pearl is single Anti-),(mycophenolic acid),(interferon beta-1a),(infliximab),(Embrel),(adalimumab),(mycophenolate) and combinations thereof, In at least one pharmaceutically acceptable carrier.
The impact of food
In certain embodiments, when experimenter is on the feed or under fasted conditions, the medicine group in embodiments of the invention Compound, such as, tablet or suspension, it is provided that to experimenter.In one embodiment, inclusion compound 12 or compound 13 The compositions of (or its pharmaceutically acceptable salt) can be supplied to fasting subject, such as, is less than 24 hours on the feed but super Spend 12 hours, more than 11 hours, more than 10 hours, more than 8 hours or more than 5 hours after.
In other embodiments, the compositions of inclusion compound 12 or compound 13 (or its pharmaceutically acceptable salt) can Or to be supplied to experimenter afterwards on the feed together with food.In one embodiment, compound 12 or compound 13 (or its medicine Acceptable salt on) can be taken by experimenter on an empty stomach.
Patients
In certain embodiments, the compound of formula (I), such as, compound 12 or compound 13 (or it is pharmaceutically acceptable Salt) (being only referred to as " compound " in this part), the compositions of inclusion compound or combination treatment be applied to about 1-6 Month, 6-12 month, 1-5 year, 5-10 year, 10-15 year, 15-20 year, 25-30 year, 30-35 year, 35-40 year, 40-45 year, 45- 50 years old, 50-55 year, 55-60 year, 60-65 year, 65-70 year, 70-75 year, 75-80 year, 80-85 year, 85-90 year, 90-95 year Or the mammal in 95-100 year.
In certain embodiments, compound, the compositions of inclusion compound or combination treatment have been administered to virus infection Dangerous people.In certain embodiments, compound, the compositions of inclusion compound or combination treatment are applied to there being virus sense The people that dye is dangerous.In certain embodiments, described patient be about 1-6 month, 6-12 month, 1-5 year, 5-10 year, 10-15 year, 15-20 year, 25-30 year, 30-35 year, 35-40 year, 40-45 year, 45-50 year, 50-55 year, 55-60 year, 60-65 year, 65- 70 years old, 70-75 year, 75-80 year, 80-85 year, 85-90 year, 90-95 year or the people in 95-100 year.
In certain embodiments, compound, the compositions of inclusion compound or combination treatment are applied to human infant.? In other embodiments, compound or combination treatment are applied to human child.In other embodiments, compound, comprise chemical combination The compositions of thing or combination treatment are applied to adult.In additionally other embodiments, the compositions of compound inclusion compound Or combination treatment is applied to old people.
All of percent used herein and ratio, except as otherwise noted, be by weight.Other feature of the present invention With advantage from different embodiments apparent.The embodiment provided illustrates the different component for implementing the present invention And methodology.Described embodiment is not intended to claimed invention.Based on present disclosure, those skilled in the art can To identify and to use other components and the methodology for implementing the present invention.
All of patent, patent application and publication mentioned above are incorporated herein by reference by entirety at this.But, Be incorporated by reference into comprises a clearly defined patent, patent application or publication, and those explicitly define and should be understood to It is applicable to be incorporated to patent, patent application or publication, wherein, finds that they are not the remainders of the text of the application, Particularly following claims.
Embodiment
Embodiment 1
Scheme 1
The synthesis of compound 3: to Me-THF (1.252ml) solution of 1-bromo-3-methybutane (156.6g, 1.037mol) Middle addition magnesium chips (30.70g, 1.263mol).After 4-5 minute, temperature starts to be increased to 50 DEG C from 21.7 DEG C.Use dry ice/ Acetone bath controls temperature.Finally, temperature is increased to 60 DEG C, then cools down.When 40 DEG C, add a fritter iodine.Then Solution is heated to 61 DEG C and stirs 2 hours.Then will add heat extraction, and mixture will be cooled to 40 DEG C.By reactant mixture It is cooled further to-59 DEG C, and in mixture, adds the Me-THF solution of the bromo-DODECANOL, 1-of 12-(50g, 0.189mol) (312.5ml), so that temperature is always not above-55 DEG C.After adding the bromo-DODECANOL, 1-of 12-, it is followed immediately by adding four The sour two lithium solution of chloro copper (II) (0.1M, 103.68ml in THF).Reactant is warmed to room temperature and stirs 16 hours.Thin Layer chromatography (TCL) (hexane: ethyl acetate is 2:1) shows that reaction completes the (R of initiation materialfIt is 0.48, the R of productfFor 0.58).Reactant is cooled to 0 DEG C and in reactant, is slowly added to saturated NH4Cl(750ml).Temperature during adding Degree is increased to 22 DEG C.By ethyl acetate (1500ml) strip aqueous.The Organic substance saline (750ml) merged is washed, MgSO4It is dried, and filters.This solution is concentrated in the vacuum of 40 DEG C, obtains the material needed for 49g (99%).This material is straight Connect for next step.
The synthesis of compound 4: to compound 3 (49g, 0.191mol) and DIPEA (DIPEA) The cold soln (0-5 DEG C) of the dichloromethane (490ml) of (27.41g, 0.212mol) is slowly added into mesyl chloride (24.29g, 0.212mol), to guarantee that temperature never can be raised to more than 5 DEG C.Reactant is warmed to room temperature, and stirs 16 hours.To instead Answer and mixture adds mesyl chloride (0.065mol, 7.4g) and DIPEA (0.52mol, 6.27g).Reactant is stirred for 24 Hour.Reactant is cooled to 6.5 DEG C, is slowly added to water (500ml).The reactant mixture of cooling is stirred 2.5 hours.Will DCM layer separates, through Na2SO4It is dried, and filters.This solution is concentrated in 40 DEG C of vacuum.100ml is added in yellow residue Methanol.This solution is stood 30 minutes at 4 DEG C, is settled out white solid.By this have precipitation solid solution filter, and Air filter is dried 16 hours.Concentrate the filtrate to half volume, wherein, be settled out other solid, then by its mistake Filter.The solid merged from above two steps is merged, in methanol (200ml), grinds 0.5 hour.Filter, be dried white Solid 16 hours, obtains the required product of 48g (75%).This material is used for next step.
The synthesis of compound 5: in 30 minutes, to the NMP's (225ml) of 1,3-PD (0.551mol, 41.96g) Cold (-5 DEG C) solution adds NaH (0.276mol, 11.02g) with aliquot (aliquot of about 0.5g).This mixture is warmed to room Temperature, stirs 30 minutes.Add in this solution and be dissolved in the compound 4 in METHYLPYRROLIDONE (NMP) (225ml) (0.135mol, 45g).Reactant is stirred 16 hours.TLC (hexane: ethyl acetate is 2:1) Indicator Reaction is complete.Initial former The R of materialfIt is 0.62, and the R of productfIt is 0.53.Water (400ml) and ethyl acetate (800ml) is added in reactant mixture.Will Organic layer separates with water layer.Organic layer is washed with water (2 × 500ml).This solution is concentrated in 40 DEG C of vacuum.By adding first Alcohol (2 × 200ml) and in 40 DEG C of vacuum concentrate mixture is further dried.The step being dried in methanol and concentrate also uses second Nitrile (2 × 200ml) repeats to obtain yellow oil.120ml acetonitrile is added in 59g yellow oil, and by this mixture Stir 1 hour at 0-5 DEG C.The waxy white solid formed in grinding from acetonitrile filters.This solid is immediately transferred into one In the round-bottomed flask of individual 500ml, and it is dried 16 hours in Rotary Evaporators, obtains 44g material.This material being dried, chemical combination Thing 5, uses in scheme 2.
Scheme 2
The synthesis of compound 8: add TMS-Br in acetonitrile (423ml) solution of compound 6 (0.146mol, 47g) (0.327mol, 50.01g), keeps the internal temperature of about 21.7-23.5 DEG C simultaneously.After the addition is complete, internal temperature is regulated To 55 DEG C, and stir 2 hours.After 2 hours, remove acetonitrile and TMS-Br, to form concentrate by the vacuum distilling at 40 DEG C. In concentrate, addition dichloromethane (423ml) is to form solution, is subsequently added oxalyl chloride (0.327mol, 41.46g), simultaneously Keep the internal temperature of 25-40 DEG C.After oxalyl chloride has added, add 2 DMF.Reactant mixture is stirred 18 hours.Will This solution is concentrated in vacuo under the external temperature of 35 DEG C.By this material (compound 7) for next step.
To the dichloromethane (423ml) of compound 5 (0.1272mol, 40g) and compound 7 (0.1462mol, 44.33g) Cold (-8 DEG C) solution in add pyridine (0.381mol, 30.18g).This reactant mixture is stirred 3 hours.TLC (hexane: second Acetoacetic ester is 2:1) show that compound 5 has been consumed.200ml water is added in reactant mixture (being cooled to 10 DEG C).Should Mixture stirs 0.5 hour.It is then peeled off organic layer.100ml water and 75ml methanol is added in organic layer.Again separate organic Layer, and concentrate in 35 DEG C of vacuum.In residue, adding 200ml acetone, and concentrating until staying thing to be dried.Add in residue Enter 200ml acetone, and with 6N NaOH (about using 15ml) by pH regulator to about 9.04.This mixture is stood at 4 DEG C 16 little Time.10g white solid precipitation is obtained after incubation period.Filter the mixture having white solid to precipitate.Add another in mixture Outer 300ml acetone.Mixture is stood 16 hours again at 4 DEG C.The brown solid obtaining significant quantity after incubation period is sunk Form sediment.Filter this mixture and be dried, obtaining the product (compound 8) of about 18g.Carry out nuclear magnetic resonance, NMR, it was demonstrated that product is compound 8.This material uses in scheme 3.Alternately prepared by compound 8 the following step (step of compound 8 with 20g scale, HPLC-ELSD purity).
Compound 4:
15-methyl-hexadecanol (1.0 equivalents, 13.4kg, 52.25mol), dichloromethane is loaded in reactor (172.2kg) and N, N-diisopropyl ethyl amine (1.3 equivalents, 8.8kg, 67.92mol).This mixture is cooled to-2.5 DEG C, And in 20 minutes, it is added thereto to mesyl chloride (1.3 equivalents, 7.8kg, 67.92mol), keep the temperature of≤2 DEG C simultaneously.? After having added, temperature regulation to 0-5 DEG C and is stirred 30 minutes.Then temperature is regulated to 20 DEG C, be stirred for 30 minutes.Enter In journey, HPLC-ELSD analyzes and determines that 15-methyl-hexadecanol has been consumed.Water (40kg) is added in reactant mixture.Will Its stirring 2 hours, then stands 0.5 hour.Separate each layer, and wash organic layer 10 minutes with water (20kg) at 21.5 DEG C. Then each layer is stood 1 hour.Separate each layer, and at a temperature of≤35 DEG C, concentrate dichloromethane until being maintained at the amount of 18L. Adding ethanol (23.7kg) in reactor, this mixture stirs 10 minutes at 20 DEG C.By mixture the temperature of≤55 DEG C Lower concentration is until keeping the amount of 18L.This ethyl alcohol azeotropy article carries out the other time.Ethanol is added in this reactor (15.8kg).Solution is warmed to 28.7 DEG C, and stirs 1 hour.Then in 37 minutes, solution is cooled to 0 DEG C.Then will It stirs at least 17 hours at 0 DEG C.Then the solid of gained is filtered.By ethanol (4.8kg) washing reactor, and it is also It is transferred in filter.Solid is transferred to pan dryer, and is dried at a temperature of≤35 DEG C, until loss on drying (LOD) it is≤1%.Obtain 16.1kg (92%).HPLC-ELSD (AUC) purity-99.3%.
Compound 5:
Reactor will load 1,3-PD (4.2 equivalents, 15.4kg, 201.6mol) and NMP (65.4kg).This is mixed Compound stirs and is cooled to-3.3 DEG C.Be dividedly in some parts in this solution in 2 hours sodium hydride (60% is dispersed in mineral oil, 2 Equivalent, 3.8kg, 96.0mol), to guarantee that temperature is maintained at≤10 DEG C.Temperature is regulated to 20 DEG C, and by this solution stirring 1 Hour.So that temperature be maintained at≤speed of 35 DEG C add in this reactor compound 4 (1 equivalent, 16.0kg, NMP (65.4kg) solution 48.1mol).The reactor being mixed with NMP (20kg) washing compound 4, it is also such that temperature Degree is maintained at≤and the speed of 35 DEG C transfers in reactor.Reaction temperature regulation to 35 DEG C and is stirred 1 hour.Then will reaction Temperature regulation to 25 DEG C and stirs 15 hours.In process, HPLC-ELSD analyzes and determines that compound 4 is consumed.To reactant mixture Middle addition heptane (35kg).Then solution is cooled to 3.6 DEG C, be added thereto in 30 minutes water (40kg) keep simultaneously≤ The temperature of 20 DEG C.Temperature regulation to 20 DEG C and is stirred 10 minutes.Water (380kg) and heptane is added in this reactor (70kg).It is stirred for 10 minutes, then stands 30 minutes.Separate each layer, and extract again with the aqueous solution of heptane (35kg).Will The Organic substance saline (11.6kg) merged stirs 10 minutes, then stands 30 minutes.Separate each layer, and through sodium sulfate (3.3kg) it is dried organic layer.It is stirred for 1 hour, then filters.With heptane (20kg) washing organic layer containing sodium sulfate Reactor, it is sent also by filter.Then concentrated solvent at a temperature of≤50 DEG C, until not observing further Distillation.In the reactor containing product, add acetonitrile (28.7kg), be heated to 45 DEG C.It is stirred for 10 minutes, so After be cooled to 0 DEG C.Then solution is stirred 1 hour at 0 DEG C.Then the solid of gained is filtered.Anti-with acetonitrile (4kg) washing Answer device, and it is also moved in filter.By this acetonitrile recrystallization again.Product is dried in the filter 1 hour.So After transfer them to pan dryer, and be dried about 72 hours at 22 DEG C, now LOD is determined as≤1.0%.Obtain 15.6kg (103.3%).HPLC-ELSD (AUC) purity is 98.3%.
Compound 7:
Compound 6 (1 equivalent, 18.8kg, 58.3mol) and acetonitrile (70.7kg) is added in reactor.To this mixture In disposably add bromotrimethylsilane (2.24 equivalents, 20.0kg, 130.6mol).Mixture is warmed to 55 DEG C and stirs 3 Hour.LC/MS determines that compound 6 is consumed.The content of this reaction is concentrated at a temperature of≤50 DEG C, until not observing To further distillation.In this reactor add 1,2-dichloroethanes (25kg), and by its at a temperature of≤50 DEG C dense Contracting, until not observing further distillation.Repeat the most once.1,2-dichloroethanes is added in this reactor (138.2kg).By temperature regulate to 20 DEG C, and be dividedly in some parts in this solution in 30 minutes oxalyl chloride (2.24 equivalents, 16.6kg, 130.6mol), maintain the temperature at≤30 DEG C simultaneously.Reaction temperature is regulated to 55 DEG C, and it is little to stir the mixture for 3 Time.31P NMR analyzes and shows that the conversion ratio of compound 7 is only 57%.In reactant mixture, dimethyl is added in 30 minutes The 1 of Methanamide (85g), 2-dichloroethanes (4kg) solution, keep the temperature at≤30 DEG C simultaneously.Then reactant mixture is existed Stir 1 hour at 25 DEG C.31P NMR analysis shows that the conversion ratio of compound 7 is 98%.By the content of this reaction at≤50 DEG C At a temperature of concentrate, until volume is at about below 40L.Addition 1 in this reactor, 2-dichloroethanes (25kg), and by it Concentrate at a temperature of≤50 DEG C, until volume is close to below 40L.By 1,2-dichloroethanes azeotropic mixture is repeated twice, and obtains The final volume of 40L.HPLC determines that purity is 90.4%.This material (compound 7) is maintained under 20 DEG C of nitrogen, until next Step (compound 8).
Compound 8:
To containing in the reactor of the solution of the compound 7 (1.25 equivalents, 17.7kg, 58.3mol) of above-mentioned steps Add compound 5 (1 equivalent, 14.7kg, 46.7mol) and 1,2-dichloroethanes (160.0kg).Then this mixture is cooled to 9 DEG C, and in 30 minutes, it is dividedly in some parts the 1 of pyridine (3 equivalents, 11.1kg, 140.1mol), 2-dichloroethanes (10kg) wherein Solution, keeps the internal temperature of≤15 DEG C simultaneously.Temperature is always not above 10.8 DEG C.Temperature regulation to 10 DEG C and is stirred 2 Hour.In process, HPLC-ELSD analyzes and determines that compound 5 has been consumed.Reactant is cooled to 3 DEG C, and in 30 minutes It is slowly added into water (3kg) wherein, keeps the temperature of≤30 DEG C simultaneously.Temperature is never higher than 19 DEG C.Ensuing 40 The water of other 67kg is added in minute.Temperature is regulated to 30 DEG C, stirs the mixture for 16 hours.Stop stirring, by each layer Stand 23 hours.Separate each layer.Water (50kg) and the mixture of methanol (40kg) is added in organic layer.It is stirred at 30 DEG C Mix 30 minutes, then stand 30 minutes.Separate each layer, with methanol/water repeated washing twice.Then in organic layer, add water (50kg) mixture of/methanol (40kg)/6N HCl (0.6kg), stirs it 30 minutes at 30 DEG C.By each layer standing sedimentation 30 minutes, it is then peeled off.Water/methanol/6N HCl washing is repeated 2 times.By organic layer≤50 DEG C of concentrations, until not observing Further distillation.Acetone (25kg) is added in the reactor containing product.By acetone≤50 DEG C of concentrations, until not having Observe further distillation.This azeotropic mixture is repeated once again.Acetone is added in the reactor containing product (100.5kg).It is added thereto to the 6N NaOH solution of 7.3kg.Add NaOH solution this amount make pH value 9.75 with On, 1N HCl solution is made, and joins in this mixture by this HCl solution of 0.4kg, to obtain the final pH of 9.68.So After acetone soln is cooled to 0 DEG C, and be added thereto to 20g compound 8 seed.It is stirred at 0 DEG C 14.5 hours, then Filter.By acetone (20kg) washing reactor at 0 DEG C, it is also channeled in filter.Then solid product is transferred to In reactor, and it is added thereto to acetone (100kg).It is stirred 1 hour at 20 DEG C, is subsequently cooled to 0 DEG C.By mixture Stir 2 hours at 0 DEG C, then filter.Again being washed with acetone (20kg) at 0 DEG C by reactor, it is also channeled into filtering In device.Product is placed and is dried under vacuum 13 hours in the filter.Product is transferred to pan dryer, and at≤30 DEG C It is dried, until LOD is≤1%.HPLC determines that purity is 95.1%.Obtain 19.3kg (70.7%).
Scheme 3
The synthesis of compound 10: (method A): load anhydrous dimethyl formamide (18kg), compound 9 in reactor (9.0kg, 1.1 work as (6kg, 1 equivalent, 14.04mol), tert-butyl alcohol magnesium (2.5kg, 1.05 equivalents, 14.74mol) and compound 8 Amount, 15.44mol).Reaction is heated to 80 DEG C and stirs 3 hours.With HPLC monitoring reaction.When compound 9 is≤20%, Reactant is cooled to 15 DEG C, and is added thereto to isopropyl acetate (56kg).(3kg HCl exists to be added thereto to HCl solution In 36kg water).Temperature is regulated to 20 DEG C, stirs the mixture for 1 hour.Stop stirring, each layer is stood 1 hour.Separate each Layer, and wash organic layer (10kg NaCl is in 40kg water) twice with saline.After final separation, 40 DEG C of vacuum remove second Isopropyl propionate, until not observing more distillation.Methanol (36.7kg) is added in reactor.By it in 40 DEG C of vacuum Middle removing, until not observing more distillation.Methanol (36.7kg) is added in reactor.This solution is directly used in down One step.
(method B): alternately, by compound 9 (0.014mol 6g), compound 8 (0.029mol, 17.17g), two uncles Butanol magnesium (0.035mol, 5.98g) adds in flask together with DMF (25ml), and is heated to 80 DEG C lasting 3 hours.Now, HPLC display reaction completes.Reactant is cooled to room temperature, is added thereto to isopropyl acetate (45ml) and 1N HCl (40ml). It is stirred for 0.5 hour.Organic layer is separated, washs with saline (2 × 41ml), through Na2SO4It is dried, and dense in 40 DEG C of vacuum Contracting.Adding methanol (2 × 100ml) in mixture, this mixture concentrates to produce compound 10 at 40 DEG C further, and it is right Afterwards for next step.
Scheme 4
The synthesis of compound 11: (method A): 0 DEG C will be cooled to from the methanol solution of previous step (method A), Xiang Qi In be passed through hydrogen chloride gas (1.6kg).This solution is warmed to 15 DEG C and stirs 2 hours.With HPLC monitoring reaction.When by chemical combination When thing 10 is≤5%, filter settled solid.This is not product.Product is in filtrate.With methanol washing (2.9kg) reaction Device, it is sent by filter.Methanol filtrate is transferred back to reactor, and is cooled to 5 DEG C.Added in this solution in 30 minutes Enter water (54.3kg), keep the temperature of≤30 DEG C simultaneously.Using pH meter, in reaction filtrate, adding 1N NaOH solution, until reaching To the pH of 2.5.Filter gained solid.Being washed by reactor use water (17.6kg), it is transferred in filter.Solid is shifted Return reactor, and be added thereto to acetone (47.2kg).It is stirred for 1 hour, filters the most under stress.Then filter cake is used Acetone (2 × 11.8kg) washs.Solid is transferred to tray dryer and is vacuum dried 12 hours at≤40 DEG C.Acetone removes After completing, solid is transferred back to reactor, and is added thereto to methanol (41kg).This mixture is heated to reflux (65 DEG C) and Stirring is until obtaining the solution of clarification.In 6 hours, solution is cooled to 0 DEG C and stirs 2 hours.Cross filter solid.At 0 DEG C By methanol (11.7kg) washing reactor, and transfer in filter.After no longer producing filtrate, solid is transferred back to reactor In, and it is added thereto to methanol (51kg).It is heated to reflux (65 DEG C) and stir until obtaining the solution of clarification.Little 6 Time interior, solution is cooled to 0 DEG C and stirs 2 hours.By filtering, solid is collected.Again with methanol (11.7kg) at 0 DEG C Washing reactor, it is also moved in filter.Then solid is transferred to exsiccator pallet the vacuum baking at≤40 DEG C Case is dried 24 hours.Obtain the compound 11 of purity >=92% of 5.6kg (69%).
(method B): alternately, at room temperature add in the compound 10 (12g, 0.0147mol) methanol (70ml) and The methanol solution (0.0441mol, 35.29ml) of 1.25N HCl.Reactant is stirred 18 hours.HPLC shows that reaction is completely.Cross Filter white solid (trityl by-product).Filtrate water (50ml) is diluted, and with 6N NaOH by pH regulator to 2.5.Should Mixture is stirred at room temperature 1 hour and filters.By product (compound 11) pulp in acetone (2 × 100ml), and filter. Product vacuum drying oven at room temperature will be dried 24 hours.Nuclear magnetic resonance, NMR confirms product.
Scheme 5
The synthesis of compound 12: (method A): in reactor load compound 11 (5.6kg, 1 equivalent, 9.73mol) and Water (84kg).It is heated to 90 DEG C and stirs 20 hours.Obtain the solution of clarification.Reactant is heated to 99 DEG C and stirs 96 Hour.HPLC determines that compound 11 is for≤1%.Reactant is cooled to 20 DEG C, and be added thereto to sodium carbonate (1.5kg, 1.5 Equivalent, 14.6mol).It is stirred for 10 minutes.Ethyl acetate (65kg)/2-propanol (6.3kg) mixing is added in this solution Thing.It is stirred for 5 minutes, and makes it separate 20 minutes.Each layer is separated.Aqueous solution (containing product) is transferred back to reactor, And it is added thereto to ethyl acetate (65kg)/2-propanol (6.3kg) mixture.It is again stirring for 5 minutes, and makes it separate 20 Minute.Each layer is separated.Aqueous solution (containing product) is transferred to reactor, and is cooled to 10 DEG C.It is carefully added into wherein HCl solution (5.8kg HCl is in 4.8kg water) keeps temperature≤25 DEG C.Temperature is adjusted to 20 DEG C, and adds in this aqueous solution Enter ethyl acetate (50.5kg)/methanol (11.1kg) solution.It is stirred for 10 minutes, and allows to separate 20 minutes.Each layer is divided From.Aqueous phase is transferred back in reactor and extracts again once with ethyl acetate (50.5kg)/methanol (11.1kg) solution.By organic Layer is transferred back to reactor, and washs with the HCl solution (1kg HCl is in 19kg water) of 0.5M.It is stirred for 5 minutes, and makes it Separate 20 minutes.Remove lower aqueous layer.HCl solution (1kg HCl is in 19kg water) and the methanol of 0.5M is added in reactor (1.6kg).It is stirred for 5 minutes, and makes it separate 20 minutes.Remove lower aqueous layer.The HCl adding 0.5M in reactor is molten Liquid (1kg HCl is in 19kg water) and methanol (1.6kg).It is stirred for 5 minutes, and makes it separate 20 minutes.Remove lower layer of water Layer.Organic substance is carried out at a temperature of≤40 DEG C vacuum distilling until not observing further distillation.To reactor Middle addition methanol (40kg), carries out vacuum distilling until not observing and further distillating at a temperature of≤40 DEG C by it Thing.Methanol (60kg) and charcoal (7kg) is added in reactor.Heat the mixture to 62 DEG C and stir 20 minutes.Then by it It is cooled to 20 DEG C and sampling carries out HPLC analysis.HPLC display purity is 97.8%.Methanol solution has been used methanol by one layer (2 × 12kg) washed kieselguhr (6kg) filters.Kieselguhr filter cake methanol (2 × 50kg) washs, to guarantee all of product Thing is all removed.Methanol containing product is transferred in reactor, and is distilled in a vacuum at a temperature of≤40 DEG C, until not having Observe further distillation.In reactor, add acetone (20kg), and distill at a temperature of≤40 DEG C, until not having Observe further distillation.In reactor, add acetone (20kg), and distill at a temperature of≤40 DEG C, until not having Observe further distillation.Acetone (20kg) is added in reactor.This solution is transferred in the rbf of a 50L. Reactor is washed with other acetone (8kg), and transfers in the rbf of 50L.Acetone soln is cooled to-45 DEG C, and stirs 20 minutes.Then product filtered and be dried.Obtain 2.13kg (38%).Purity >=97%.
(method B): alternately, adds water (90ml) in compound 11 (0.01563mol, 9g).Mixture is heated To 90 DEG C and stir with mechanical agitator.After 1 week, HPLC shows to react and has completed more than 95%.Mixture is cooled to room temperature. PH to 1-2 is regulated with 1N HCl.With ethyl acetate (3 × 500ml) extraction water solution.The acetic acid ethyl ester extract warp merged Na2SO4It is dried, filters, and be concentrated in vacuo at 40 DEG C, obtain the thick grease of 9g.Used acetone (100ml) to grind, and cooled down To 4 DEG C.After white solid filtration drying, obtain 5g product.NMR confirms that this product is compound 12.
Embodiment 2
Antiviral and cytotoxicity experiment
The EC of the human cytomegalovirus (HCMV) in MRC-5 cell50: Costar 96 hole tissue culturing plate is existed Inoculating 20000MRC-5 cells/well in EMEM, described EMEM contains 2% hyclone standard hyclone and 1% hyclone Penicillin and streptomycin.Exit orifice is not used in and minimizes by extending the edge effect that incubation produces.Cell with 0.01 the HCMV of MOI Inoculation.By test compound serial dilution join in cell, plate 5% CO2In 37 DEG C of incubations 7 days.7 days temperature After educating, Positive control wells shows the cellular morphology instruction that the HCMV in 90-100%MRC-5 cell infects.After 7 days incubations, will Culture medium is removed lightly from the cell infected.Being rinsed twice with ice-cold PBS by cell, then freeze/thaw is once.Respectively Hole at 55 DEG C with 200 μ L lysis buffer incubation 2 hours.Lysis buffer includes the E.C. 3.4.21.64 of 0.5mg/mL, 50mM KCl, pH are 10mM Tris-Cl, the 2.5mM MgCl of 8.02, the IGEPAL of 0.45% and be dissolved in DEPC and process in water 0.45% tween 20.Intracellular CMV DNA is by using forward and reverse HCMV PCR primer (qPCR) and FAM labelling The quantitative polyase chain reaction of probe measures.Use the HCMV DNA cloning used containing with the sequence of amplified fragments homology The standard curve of the diluent of son carries out the absolute quantitation of virus replication number.Following qPCR amplification condition is used: 1 circulation Carry out 10 minutes at 95 DEG C, be that 45 circulations of 95 DEG C carry out carrying out 60 seconds in 15 seconds and 60 DEG C subsequently.QPCR reaction should with the U.S. Carry out with 7500 real-time PCR systems of Biosys Corp..5th generation software (Bai Teng Instrument Ltd. of the U.S.) is used for calculating pressing down The viral DNA levels of the MRC-5 cell that HCMV processed infects is to 50% (EC50) concentration.
The EC of the BKV in VERO cell50: Costar 96 hole tissue culturing plate is inoculated 10000Vero in DMEM thin Born of the same parents/hole, described DMEM contains 2% hyclone standard hyclone (FBS, Cat SH30088.03) and 1% hyclone is blue or green Mycin and streptomycin.Exit orifice is not used in and minimizes by extending the edge effect that incubation produces.Cell 115BKV DNA copy/thin Born of the same parents' (ATCC, Gardner's strain) inoculate.By test compound serial dilution join in cell, plate 5% CO2In 37 DEG C incubation 10 days.After 10 days incubations, by 50 μ L 2X of the supernatant of 50 μ L with the E.C. 3.4.21.64 providing final concentration 0.5mg/mL Lysis buffer, the 10mM Tris-Cl that KCl, pH are 8.0 of 50mM, 2.5mM MgCl2, the IGEPAL of 0.45% and being dissolved in DEPC processes 0.45% tween 20 mixing in water.By each plate incubation 2 hours at 55 DEG C.The BKV DNA of supernatant is by making Measure with the quantitative polyase chain reaction (qPCR) of forward and reverse BKV PCR primer and FAM label probe.Use is adopted The exhausted of virus replication number is carried out with containing the standard curve with the diluent of BKV DNA cloning of the sequence of amplified fragments homology To quantitatively.Following qPCR amplification condition is used: 1 circulates in 95 DEG C and carries out 10 minutes, is 45 circulations of 95 DEG C subsequently Carry out carrying out 60 seconds in 15 seconds and 60 DEG C.QPCR reaction is carried out with 7500 real-time PCR systems of Applied biosystems.The 5 generation softwares (Bai Teng Instrument Ltd. of the U.S.) are used for calculating the viral DNA levels of the Vero cell that suppression BKV infects to 50% (EC50) concentration.
Cytotoxicity (CC in Vero cell50): by Costar 96 hole tissue culturing plate at DMEM (ACTT, Cat Inoculation 10000Vero cell (ATCC)/hole in 30-2002), described DMEM contain 2% hyclone standard hyclone (FBS, Cat SH30088.03) and 1% hyclone penicillin and streptomycin (Cat SV30010).Exit orifice is not used in and minimizes by prolonging The edge effect that long incubation produces.By test compound serial dilution join in cell, plate 5% CO2In 37 DEG C incubation 7 days.After the incubation of 7 days, the culture medium by manufacturer's instruction 200 μ L in each hole adds 40 μ L cells and drips DegreeThe MTS reagent (Promega company, G111) of aqueous solution, and 37 DEG C of cultivations, until untreated cell controls is formed The absorbance between 1.1 and 1.8 at 490nm.Final absorbance reading uses Bai Teng Instrument Ltd. of the U.S. Synergy2, and use the 5th generation software (Bai Teng Instrument Ltd. of the U.S.) to calculate suppression Vero cell to 50% (CC50) Concentration.
Cytotoxicity (CC in MRC-5 cell50): by Costar 96 hole tissue culturing plate at EMEM (ACTT, Cat Inoculation 20000MRC-5 cell (ATCC)/hole in 30-2003), described EMEM contains 2% hyclone standard hyclone (FBS, Cat SH30088.03) and 1% hyclone penicillin and streptomycin (Cat SV30010).Exit orifice is not used in and minimizes By extending the edge effect that incubation produces.By test compound serial dilution join in cell, plate 5% CO2In 37 DEG C of incubations 7 days.After the incubation of 7 days, the culture medium by manufacturer's instruction 200 μ L in each hole adds 40 μ L thin Born of the same parents' titreThe MTS reagent (Promega company, G111) of aqueous solution, and 37 DEG C of cultivations, until untreated cell controls It is formed at the absorbance between 1.1 and 1.8 at 490nm.Final absorbance reading uses Bai Teng Instrument Ltd. of the U.S. Synergy2, and use the 5th generation software (Bai Teng Instrument Ltd. of the U.S.) calculate suppression MRC-5 cell to 50% (CC50) concentration.
Cytotoxicity (CC in MT4 cell50): by Costar 96 hole tissue culturing plate at RPMI (Lonza, Cat Inoculation 5000MT4 cell (NIH acquired immune deficiency syndrome (AIDS) Reagent Project)/hole in 12-11F), described RPMI contains 10% hyclone standard tire Ox blood serum (FBS, Cat SH30088.03), 1% hyclone penicillin and streptomycin (Cat SV30010) and the Lonza of 2mM L-glutaminate (Cat 17-605E).Exit orifice is not used in and minimizes by extending the edge effect that incubation produces.Compound will be tested Serial dilution join in cell, plate 5% CO2In 37 DEG C of incubations 6 days.After the incubation of 6 days, refer to by manufacturer Show and the culture medium of 200 μ L in each hole adds 40 μ L cell titersThe MTS reagent of aqueous solution (Promega company, G111), and 37 DEG C of cultivations, until untreated cell controls is formed at the absorbance that 490nm is between 1.1 and 1.8.? Whole absorbance reading uses the Synergy2 of Bai Teng Instrument Ltd. of the U.S., and (U.S. uncle rises instrument to be had to use the 5th generation software Limit company) calculate suppression MT4 cell to 50% (CC50) concentration.
Cell is cultivated and Strain: is organized in the University of Alabama of buying equipment its IRB approval from Birmingham and obtains People's prepuce tissues prepare human foreskin fibroblast (HFF) cell.This is organized at 4 DEG C incubation 4 in cell culture medium Hour, described cell culture medium is put down by the ell being supplemented with 10% hyclone (FBS) (Hyclone company, Lip river root UT) MEM (MEM) composition of weighing apparatus salt and the glutamine of normal concentration, amphotericin B and vancomycin.Then will Tissue is placed in phosphate buffered saline (PBS) (PBS), chopping, cleans and removes Red blood corpuscle, and is resuspended in trypsin/EDTA In solution.By suspensions of tissues in 37 DEG C of incubations, and it is gently mixed with cell dispersion, is then collected by centrifugal.By cell weight It is suspended in 4mL culture medium, and is placed on 25cm2In tissue culture flasks, at moistening CO2In incubator, at 37 DEG C of incubations 24 Hour.Then culture medium fresh culture being substituted, monitoring cell growth every day until forming the cell monolayer merged.Then By the HFF cell standard by the MEM at E Ershi salt, L-glutaminate, penicillin and the gentamycin being supplemented with 10%FBS In growth medium, continuous passage expands.Cell routine is passed on, and will be used for measuring equal to or less than 10 generation cells.
Vero cell is from American type culture collection (American Type Culture Collection) (ATCC, Manassas, VA) obtains, and maintains E Ershi salt, L-glutaminate, penicillin and the chain being supplemented with 10%FBS In the standard growing media of the MEM of mycin.
The A Kata cell of latent infection EBV obtains from John Sixbey.Studied and reference reagent by NIH acquired immune deficiency syndrome (AIDS) Plan obtains the GS strain of HHV-6A.
Antiviral breeding: each experiment of the antiviral activity evaluating compound includes positive and negative control chemical combination Thing, to guarantee the performance of each mensuration.Each research of the peer-level of compound of coming into the open is also carried out Cytotoxic parallel Assessment.When there being enough materials can use, it is respectively directed to every kind of compound and carries out multiple analysis and evaluation acquisition statistical data.
The plaque subtrahend experiment of HSV-1, VZV and HCMV: prepare HFF cell monolayer in 6 orifice plates, and at 37 DEG C of incubations 2 My god, so that cell reaches to merge.Then by culture medium sucking-off from hole, and in three holes, the virus of 0.2mL is added, with often Individual hole obtains 20-30 speckle.Virus is allowed to be adsorbed onto cell 1 hour, and every 15 minutes jiggle plate with again Distribution culture medium.Compound is diluted in and comprises that to be supplemented with E Ershi salt, L-glutaminate, penicillin and the celebrating of 2%FBS the most mould In the maintenance cell culture medium of the MEM of element.According to virus used, to replicating add from 300 μMs to 0.1 μM scopes in hole molten Liquid, and repeatedly plate described in incubation.The plaque subtrahend experiment of HSV-1 bacterial strain F is carried out in a similar manner, but uses coated plate (plating) the Vero cell infected one day after.The most final FBS concentration is 5%.For HSV-1 and-2, monolayer 1% violet staining being used in 20% methanol, unconjugated dyestuff passes through dH2O washs removing.HCMV and VZV is tested, Cell monolayer dyes 4 hours by 1% neutral red solution, is then aspirated by dyeing liquor, by cells rinsed with PBS.For all of Experiment, uses stereoscopic microscope calculate plaque and reduce by 50% (EC from quality evaluation of the experimental data50) speckle formed compound Concentration.
The DNA hybridization experiment of EBV, HHV-6A and HHV-6B.EBV experiment is being induced to experience 50 μ g/ by standard method The A Kata cell of the lytic infection of goat anti-human immunoglobulin's antibody of ml is carried out.In round bottom 96 orifice plate, dilution is real Test compound to produce the concentration from 20 to 0.016 μM of scope.With every hole 4 × 10 in described plate4The concentration of individual cell adds A Kata cell also cultivates 72 hours.HHV-6 is tested, serial dilution compound in 96 orifice plates, in each hole, then add 1 ×104HSB-2 or the Molt-3 cell being uninfected by.By every 10 are uninfected by with nearly 1 cell infected respectively HSB-2 cell or the ratio of Molt-3 cell, add and infected the HHV-6A cell of HSB-2 or infected the HHV-6B of Molt-3 Cell starts to infect, and cultivates 7 days at 37 DEG C.
For all of experiment, in each hole, add 100 μ L denaturation buffer (1.2M NaOH, 4.5M 80 NaCl), With denatured DNA, use biodot device (Bio-Rad company, Heracles (Hercules), CA) by Immobilon Buddhist nun Dragon film (Mi Libo (Millipore), Bedford (Bedford), Massachusetts) aspirates 50 μ L aliquot.Then this film is permitted Permitted at digoxin simplicity hybridization solution (DIG Easy Hyb) (Roche Diagnistics (Roche Diagnostics), Indiana Pohle This, the state of Indiana) balance before at 56 DEG C be dried 30 minutes.Experimental technique (Roche Diagnistics) according to manufacturer is prepared for The probe of specifically Gaoxin (DIG) labelling of each virus.For EBV, primer 5 '-CCC AGG AGT CCC AGT AGT CA-3 ' and 5 '-CAG TTC CTC GCC TTA GGT TG-3 expands corresponding to (AJ507799) coordinate in EBV genome The fragment of 96802-97234.Probe the primer 5 '-CCT TGA TCA TTC GAC of specific HHV-6 digoxigenin labeled Fragment (in the X83413 coordinate of CGT TT-3 ' and 5 '-TGG GAT TGG GAT TAG AGC TG-3 ' amplification ORF2 37820-38418) prepare.By the hybridized overnight at 56 DEG C of the film containing EBV DNA, the most at that same temperature, with containing 0.2 × the SSC of 0.1%SDS and washing successively with 0.1 × SSC containing 0.1%SDS.To HHV-6A and HHV-6B trace, it is allowed to Probe is at 42 DEG C of hybridized overnight, at the same temperature with 0.2 × SSC containing 0.1%SDS with 0.1 × SSC containing 0.1%SDS Rinsing trace.The experimental technique (Roche Diagnistics) using manufacturer carries out the inspection of specific binding DIG probe with anti-dig antibody Survey.Capture the image of film and quantify with QuantityOne software (Bio-Rad company), enough by quality evaluation of the experimental data The accumulation of viral DNA reduces to 50% (EC50) the concentration of compound.
(v) influenza virus
Experiment based on cell.For dose-effect curve, to each used concentration, use three holes in 96 holes Microwell plate (8 × 104Cells/well) in mdck cell in add single medicine.Compound is added: Austria takes charge of under following concentration His Wei carboxylate is at 0,0.000032,0.0001,0.00032,0.001,0.0032,0.01,0.032,0.1,1.0,10.0 and 100μg/ml;Amantadine and ribavirin are at 0,0.001,0.0032,0.01,0.032,0.1,0.32,1,3.2,10,32 and 100μg/ml.Each bread board includes the untreated of the cell (virus control) that infects and the cell (cell controls) that is uninfected by Hole.After infecting at three days, virus control wells shows the cytopathology of 100%.The journey of viral cytopathic in each hole Degree is by observing with microscope and determining with dimethyl diaminophenazine chloride (NR) dyeing.In brief, by cell with being diluted in MEM 0.011%NR dyes to determine cell viability.After two hours, being processed by plate, the NR that picked-up quantifies imports living cells.By carefully Born of the same parents absorb the amount spectrophotometry of NR.
Cytotoxicity experiment: each Antiviral breeding includes using for the identical cell of each virus, identical thin Born of the same parents' quantity, identical drug level provide identical medicament contact with the identical parallel cell toxicity test cultivating the time. Cytotoxicity in order to ensure all of compound can directly compare, and we are also to the fusion HFF cell at the culture period of 7 days In all compounds carried out standard Neutral red picked-up cytotoxicity experiment.
(i) red untake cytotoxicity experiment.Under standard cytotoxic is tested, each chemical combination is evaluated by standard method Thing.In short, by HFF cell in standard tissue culture with 2.5 × 104Individual cells/well is seeded in 96 hole tissue culturing plates In.After cultivating 24 hours, culture medium with maintain cell culture medium substitute, compound is joined the first row, then 5 times continuous dilute Release liquid and be subsequently used for producing the concentration of a series of compound with most 300 μMs.Brassboard is cultivated 7 days, to each Kong Zhongjia Enter the PBS solution of the dimethyl diaminophenazine chloride of the l0.66mg/ml of 100 μ l, and this plate is cultivated 1 hour.Then coloring agent is removed, use PBS Rinse described plate, will be dissolved in by the dyestuff of living cells internalization in the PBS being supplemented with 50% ethanol and 1% glacial acetic acid.Then exist 550nm measures optical density, and from quality evaluation of the experimental data CC50Value.
All plaque subtrahends in HFF cell are tested, receives for the identical chemical combination of Antiviral breeding comprising Parallel group of 6 orifice plates of the HFF cell being uninfected by of the concentration of thing carry out dimethyl diaminophenazine chloride cytotoxicity experiment.As each antiviral is real Testing, take out cytotoxicity plate on the same day from incubator, cell monolayer 2ml concentration in PBS is the neutrality of 0.165mg/ml Red solution dyes 6 hours.Then this dyestuff is removed, the PBS of residual dye in cell is rinsed, visual inspection cell monolayer The toxicity of any sign.Vero cytotoxicity experiment is carried out to the drug level of 1mM scope with from 1 μM.Saying according to manufacturer Bright use cell titerAqueous list Solution Cell Proliferation experiment (Promega company) measures cell viability.
(ii) cytotoxicity experiment in lymphocyte.Use cell titer globulin luminescent cell activity experiment (Promega company) assessment lymphocyte cell viability in all experiments.In short, brassboard at room temperature to be cultivated 30 Minute, in each hole, then add the cell titer globulin reagent of 50 μ l, and plate is mixed in orbital shaker 2 minutes with Cell lysis.Then plate is at room temperature cultivated other 10 minutes, and quantitative luminescence in photometer.Calculate with standard method and press down A Kata processed (Akata), HSB-2, BCLB-1 or Molt-3 cell proliferation are to 50% (CC50) drug level.
(iii) cell proliferation experiment.The suppression of HFF cell proliferation is used to improve Cytotoxic to some compounds Estimate, and carry out according to the standardization program used in the lab.With 2.5 × 104Individual cells/well and standard medium are by cell It is inoculated in 6 orifice plates with low-density.After 24 hours, by culture medium sucking-off, and start preparation growth medium from 300 μMs Compound solution scope, and join in duplication hole.Flat board is cultivated 72 hours at 37 DEG C, then with trypsin, cell is taken off Fall, and count in Beckman Coulter-counter.The compound concentration of 50% it is reduced to from quality evaluation of the experimental data cell proliferation.
Table 2 is compound 12 anti-herpes simplex virus 1 (E-377 strain) and HCMV (AD169 in human foreskin fibroblast Strain) activity
Table 3 is compound 12 and hexadecane propyl group cidofovir (HDP-CDV) anti-HCMV in human foreskin fibroblast The activity of UL54 drug resistant mutants
Table 4 is compound 12 anti-vaccinia virus (Copenhagen strain) and vaccinia virus (Bu Lai in human foreskin fibroblast Strain) activity
Table 5 is the activity of compound 12 resisiting influenza virus strain in Madin-Darby Testis et Pentis Canis (MDCK) cell
Table 6 is the activity of the anti-epstein-Barr virus of compound 12 in A Kata cell
Table 7 compound 12 and HDP-CDV in MRC-5 cell anti-HCMV and in Vero cell the activity of anti-BKV
Table 8 compound 12 antiviral activity in vitro
Table 9 external activity contrast (meansigma methods XC50)
In a word, these data show that compound 12 has the curative effect/toxicity ratio of improvement.UL54 HCMV resistant variants's number Reduce the curative effect of compound 12 according to the sudden change showing this gene, it also reduces the curative effect of HDP-CDV, and this shows compound 12 It is destroyed as triphosphoric acid equivalent, and as the substrate inhibitor of a kind of replacement for CMV polymerase UL54.
The clinical research of embodiment 3:CMV
Compound 12 and the clinical research of compound 13 are carried out.Placebo has been carried out in HSCT CMV (R+) receiver Comparison, the experiment of dose-escalating, evaluate compound 12 or compound 13 prevents or controls the ability of cmv infection.Set up several Queue, wherein participant or experimenter are administered orally and accept placebo or compound 13, and dosage range is from 40-1000mg daily or weekly (QW) 200mg or 100mg twice a week to 40-1000mg (BIW) twice a week, such as, is accepted once in a week.Being subject to after HSCT Examination person is registered implanting when, is probabilistically assigned compound 13 or placebo (ratio of 3:1), and it is straight to accept the treatment of blind property To transplant after about 100 days.The dosage of compound 12 or compound 13 is at 40-1000mg daily or weekly with 40-1000mg weekly Twice, such as, daily or weekly, daily or weekly, daily or weekly, 200mg is twice a week and 100mg for 200mg for 100mg for 40mg Twice a week.Suffer from the experimenter first sending out treatment of medical care standard of CMV disease or cmv infection needs locality all from blind Sex therapy Stop, and follow up a case by regular visits to 4 weeks.Complete the experimenter after the treatment of blind Sex therapy and follow up a case by regular visits to 8 weeks after the treatment.
Patient accepts drugs 9-11 week, this that day randomly choosed after depending on transplanting, so that this drugs Stopping in the 13rd week after transplanting in the patient complete drugs.While patient accepts drugs, by center Laboratory carries out the measurement weekly of the plasma C MV DNA level by PCR experiment.If CMV disease development or if as In blood plasma, CMV DNA detected or because other reasons patient needs the Drug therapy with anti-cmv infection, stop drugs, And according to the practical situation treatment patient that research is on-the-spot.
Primary efficacy endpoint is to stop Advancement Type cmv infection, and it is defined as CMV disease or plasma C MV DNA level More than every milliliter of 200 copy, detect in central laboratory in 1 week after the final dose of drugs.If patient exists Research end has 200 every milliliter of copies or less plasma C MV DNA level and does not have the CMV disease of confirmation, then study Treatment (with compound 12 or compound 13 or placebo) is considered as successful, even if it is specific to use period at drugs Measure weekly and be greater than 200 parts every milliliter, the most again decline.If patient interrupts drugs, to start for cmv infection Or the treatment of other reasons, but plasma C MV DNA level is 200 every milliliter of copies or following, and CMV disease is not by really Recognize, then be considered as successful with the treatment of compound 12 or compound 13.Preassigned secondary endpoints includes cmv infection Generation or be negative or positive patient to CMV DNA at baseline (first day either used in examination or drugs) Plasma C MV DNA level increase, drugs stopping rate and stop reason, treatment CMV reaction antiviral agent use and Compound 12 or compound 13 and the trough level of cidofovir.Safety endpoints includes all untoward reaction, experiment value and the heart The death that the change of electrograph assessment and any reason cause.
The clinical research of embodiment 4:BKV
Random, the placebo pair in the 9-11 week that the BKV of HCT infects after having carried out compound 12 or compound 13 prevention According to, double blinding, dosage escalation clinical research (40-1000mg is weekly and 40-1000mg twice a week, such as, 40mg is weekly Once, 100mg is weekly, and 200mg is weekly, and 200mg is with 100mg twice a week twice a week).Treat and open when transplanting Begin and be continued until rear HCT the 13rd week.
The research of the end organ damage relevant with BKV
Analyze sickness rate and the impact of end organ complications that the experimenter BKV in queue infects.If viruria Disease exists, and measures BK virus urine disease every time and assesses viremia.Analysis and research data are with assessment compound 12 or compound 13 Whether BKV is infected end-organ disease to have an impact.Microscopic hematuria is defined as confirming haemachrome positive urine analysis;Renal function damages Wound is defined as last measurement during treating and has the kreatinin (>=120 μm ol/L) of rising, and this is also to increase from baseline Add >=25%.
In clinical studies, participate in research experimenter in, some experimenters with different dosage accept placebo and Other experimenters accept compound 12 or compound 13 with different dosage.
Definition and method
The end-organ impact of clinical meaning is defined as follows:
Microscopic hematuria is at least 1+ haemachrome labelling in urinalysis (reagent paper dipping)
New send out hematuria at least 1+ haemachrome (pass through >=trace measure confirmation continuously), only occur during treating
Serum creatinine >=120 μM (>=1.36mg/dl) at the end of renal dysfunction treatment at the end for the treatment of
Newly send out serum creatinine >=120 μM (>=1.36mg/dl) at the end of renal dysfunction treatment and at least than base Line serum creatinine big 25%
According to treatment group (comparing with placebo, amalgamation compound 13) and BKV state (any time during treating Viruria positive or negative) experimenter is listed.Use Fisher accurately to check to compare two-by-two.Due to limited sample Product size, compares with placebo group, the data of compound 12 is put together.
In order to probe into the impact of the immediate symptoms that BKV infects, before dosing (that is, the PCR of BKV is positive at baseline), first Analyze the sickness rate of the AEs comprising term BKV urine in the experimenter that BKV infects.
In order to inquire into the compound 12 impact on acute hemorrhage cystitis further, probe into the morbidity of acute hemorrhage treatment Rate.
Equivalent
The present invention can with other without departing from their spirit or basic feature concrete form embody.Therefore, previous embodiment It is considered to be illustrative rather than in all respects limiting invention as described herein.The scope of the present invention is therefore by appended power Profit claim rather than represented by description above, and the changed purport in the full scope of equivalents of implication and claims It is being included therein.

Claims (41)

1. one kind by the compound as shown in following formula (I):
Wherein, R is
Compound the most according to claim 1, it is characterised in that R is:
3. compound or its pharmaceutically acceptable salt, the choosing of described compound is freely
The group of composition.
4. a synthesis typeThe method of compound, wherein, institute The method of stating comprises the steps:
I () adds magnesium in 2-methyltetrahydrofuran (Me-THF) solution of bromo-3 methybutanes of 1-, be subsequently heated, then cool down This mixture;
(ii) in the reactant mixture of step (i), add the Me-THF solution of the bromo-DODECANOL, 1-of 12-, add immediately after Oxolane (THF) solution of four chloro copper acid two lithiums;
(iii) with the aqueous phase in ethyl acetate back extraction reactant mixture;
(iv) by saline washing organic solution and through MgSO4It is dried, filters, concentrate the most in a vacuum to produce compound 3;
V () adds mesyl chloride in the cold soln of compound 3 and the dichloromethane of DIPEA (DIPEA), with Time maintain the temperature at less than about 5 DEG C;
(vi) make reaction warm to room temperature and stir several hours;
(vii) in reactant mixture, add mesyl chloride and DIPEA and stir several hours;
(viii) add water while cooling reactant mixture, from water layer, then separate dichloromethane (DCM) layer, drying agent It is dried, and filters DCM layer to remove desiccant;
(ix) DCM solution it is concentrated under vacuum to obtain yellow oil;Methanol is added in the concentrate of yellow oil;
X the solid of precipitation is filtered and is dried by (), to produce compound 4;
(xi) adding sodium hydride (NaH) in the cold soln of the METHYLPYRROLIDONE (NMP) of 1,3-PD, warm should Mixture;
(xii) compound 4 is joined in the solution of step (xi), stir several hours;
(xiii) in this solution, add water and ethyl acetate, separate organic layer;
(xiv) concentration of organic layers in a vacuum, adds methanol, is then dried;
(xv) add acetonitrile, repeat step (xiv), after filtration and drying, form compound 5;
(xvi) in the acetonitrile solution of compound 6, bromotrimethylsilane (TMS-Br) is added;
(xvii) add dichloromethane after removing acetonitrile and TMS-Br, then concentrate;
(xviii) add oxalyl chloride and dimethylformamide (DMF), concentrate in a vacuum and form compound 7;
(xix) mixing cpd 5 and compound 7 in dichloromethane, and in the solution of compound 7 and compound 5, add pyrrole Pyridine;
(xx) organic layer is separated after the mixture in step (xix) adding water;
(xxi) organic layer is again separated after the organic layer in step (xx) adding water and methanol;
(xxii) it is dried in a vacuum the organic layer of step (xxi), and adds acetone;
(xxiii) it is dried and before being further dried, adds acetone, regulating pH to about 9.0;
(xxiv) acetone is added after the solid formed in filtration step (xxiii);
(xxv) solid product of also drying steps (xxiv) is filtered, to produce compound 8;
(xxvi) mixing cpd 8, compound 9, two-tert-butyl alcohol magnesium in DMF;
(xxvii) in mixture, add isopropanol and hydrochloric acid (HCl), be then peeled off organic layer;
(xxviii) concentration of organic layers, and in mixture, add methanol;
(xxix) mixture of concentration step (xxviii), to produce compound 10;
(xxx) in compound 10, add methanol, and filter substituent group product;
(xxxi) filtrate in dilute with water step (xxx), pH is to about 2-3 in regulation, and filters to produce compound 11;
(xxxii) in compound 11, add water, and regulate pH to about 1-2 with HCl;
(xxxiii) aqueous solution of step (xxxii) it is extracted with ethyl acetate;
(xxxiv) acetic acid ethyl ester extract of drying steps (xxxiv), filters and concentrates to produce viscous crude;With
(xxxv) grind described viscous crude with acetone, this solution is cooled down, filter and be dried, to obtain the chemical combination as white solid Thing 12.
5. the compound described in claim 1 or a pharmaceutical preparation for its pharmaceutically acceptable salt, it comprises carrier.
Pharmaceutical preparation the most according to claim 5, it is characterised in that comprise the compound of the formula (I) of about 40-1000mg.
Pharmaceutical preparation the most according to claim 6, it is characterised in that comprise the compound of the formula (I) of about 40-400mg.
Pharmaceutical preparation the most according to claim 7, it is characterised in that comprise the compound of the formula (I) of about 100mg.
9. treating virus infection and/or the disease relevant with virus infection or a method for disease, it includes in need The compound of the claim 1 of experimenter's administering therapeutic effective dose or its pharmaceutically acceptable salt.
10. treating virus infection and/or the disease relevant with virus infection or a method for disease, it includes in need The pharmaceutical preparation of the claim 5 of experimenter's administering therapeutic effective dose.
11. methods according to claim 9, it is characterised in that the amount of application of the compound of claim 1 is at about 40- In the range of 1000mg.
12. methods according to claim 10, it is characterised in that described pharmaceutical preparation includes the amount of about 40-1000mg The compound of claim 1.
13. methods according to claim 9, it is characterised in that the compound of claim 1 is executed with the amount of about 40-400mg With.
14. methods according to claim 9, it is characterised in that the compound of claim 1 is used with the amount of about 100mg.
15. 1 kinds delay morbidity in the experimenter infected BK virus, reduce risk or treatment end organ damage or damage Method, described method includes to the Orally administered pharmaceutical composition of experimenter, described pharmaceutical composition include treat effective dose The compound selected from claim 1 in any one compound or its pharmaceutically acceptable salt.
16. methods according to claim 15, it is characterised in that after described experimenter is hematopoietic stem cell transplantation (HSCT) Experimenter.
17. methods according to claim 15, it is characterised in that described end-organ is selected from kidney, ureter, bladder, front Row gland and urethra.
18. methods according to claim 9, it is characterised in that with the described compound every day of about 40-1000mg, weekly Once (QW) or twice a week (BIW) treat described experimenter's quilt.
19. 1 kinds are having BK virus to infect reduction hematuria or the method for renal damage incidence rate in the experimenter that reactivation is dangerous, should Method includes any one change in the Orally administered compound selected from claim 1 comprising treatment effective dose of experimenter Compound or the pharmaceutical composition of its pharmaceutically acceptable salt.
20. methods according to claim 19, it is characterised in that described experimenter is experimenter after HSCT.
21. methods according to claim 19, it is characterised in that described pharmaceutical composition reduces described experimenter further In BK virus infect reactivation.
22. methods according to claim 21, it is characterised in that described pharmaceutical composition reduces the BK in described experimenter Virus load.
23. methods according to claim 22, it is characterised in that described pharmaceutical composition delays end organ damage or damage The morbidity of wound or reduce end organ damage or the danger of damage, wherein, described end-organ is selected from kidney, ureter, bladder, front Row gland and urethra.
24. methods according to claim 22, it is characterised in that described experimenter every day, weekly (QW) or two weeks Once (BIW) is applied the described compound of about 40-1000mg.
25. methods according to claim 22, it is characterised in that the 1st day, the 2nd day or the 4th day after after HSCT to Described experimenter uses 1.25mg/kg, 2.5mg/kg, 5.0mg/kg, 10.0mg/kg or 20.0mg/kg.
26. 1 kinds of treatments, prevent or delay hepatitis C virus (HCV) to infect or the disease relevant with HCV infection or disease are sent out Sick method, described method includes to the Orally administered compound selected from claim 1 comprising treatment effective dose of experimenter In any one compound or the pharmaceutical composition of its pharmaceutically acceptable salt.
27. methods according to claim 26, it is characterised in that every day, weekly (QW) or biweekly (BIW) use Experimenter described in the described compounds for treating of about 40-1000mg.
28. methods according to claim 27, it is characterised in that every day or biweekly (BIW) are with about 40-1000mg's Experimenter described in described compounds for treating.
29. 1 kinds of prophylactic treatments, prevent or delay hepatitis C virus (HCV) to infect or the disease relevant with HCV infection or The method of disease morbidity, described method include to experimenter Orally administered comprise treatment effective dose selected from claim 1 Any one compound in compound or the pharmaceutical composition of its pharmaceutically acceptable salt.
30. methods according to claim 29, it is characterised in that every day, weekly (QW) or biweekly (BIW) use Experimenter described in the described compounds for treating of about 40-1000mg.
31. methods according to claim 30, it is characterised in that every day or weekly (BIW) are with about 40-1000mg's Experimenter described in described compounds for treating.
32. 1 kinds of treatments, prevent or delay hepatitis C virus (HCV) to infect or the disease relevant with HCV infection or disease are sent out Sick method, described method includes to the Orally administered compound selected from claim 1 comprising treatment effective dose of experimenter In any one compound or the pharmaceutical composition of its pharmaceutically acceptable salt, and selected from immunosuppressant and antiviral One or more compounds of agent or compositions.
33. methods according to claim 32, it is characterised in that described pharmaceutical composition with selected from by midazolam, Ciclosporin A, tacrolimus, azole, ganciclovir, valganciclovir, foscarnet sodium, cidofovir, two wires anti-HCV medicament, FOSCARNET, vein (IV) injection cidofovir, filgrastim, Polyethylene Glycol filgrastim, corticosteroid such as budesonide, One or more compounds or compositions in the group that beclometasone and wide spectrum CYP inhibitor amino benzotriazole are constituted combine and execute With.
34. according to the method described in any one in claim 9,10,15,26,30 and 32, it is characterised in that described tested Person is the experimenter of immunologic hypofunction.
35. methods according to claim 34, it is characterised in that the experimenter of described immunologic hypofunction is immunosuppressant The transplant patient of medicine.
36. methods according to claim 34, it is characterised in that the experimenter of described immunologic hypofunction has infected HIV.
37. methods according to claim 34, it is characterised in that other the immunity of described compound and at least one presses down Preparation combines and uses.
38. according to the method described in claim 37, it is characterised in that described immunosuppressant is applied simultaneously or sequentially.
39. according to the method described in claim 37, it is characterised in that described immunosuppressant selected from by daclizumab, Basiliximab, tacrolimus, sirolimus, mycophenolate, Ciclosporin A, glucocorticoid, CD 3-resisting monoclonal antibody, anti- Anti-thymocyte globulin, anti-CD52 monoclonal antibody, azathioprine, everolimus, dactinomycin, cyclophosphamide, platinum, nitroso-group Urea, methotrexate, mercaptopurine, not Luo Dankang, interferon gamma, infliximab, Embrel, adalimumab, his pearl The group that monoclonal antibody, FTY720 and combinations thereof are constituted.
40. pharmaceutical preparatioies according to claim 5, it is characterised in that described preparation is applicable to Orally administered.
Treating virus infection and/or the disease relevant with virus infection or the method for disease for 41. 1 kinds, it includes in need The pharmaceutical preparation of the claim 40 of experimenter's administering therapeutic effective dose.
CN201480074587.9A 2013-12-05 2014-12-05 Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof Pending CN106061982A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361912407P 2013-12-05 2013-12-05
US61/912,407 2013-12-05
PCT/US2014/068932 WO2015085256A1 (en) 2013-12-05 2014-12-05 Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof

Publications (1)

Publication Number Publication Date
CN106061982A true CN106061982A (en) 2016-10-26

Family

ID=53274194

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201480074587.9A Pending CN106061982A (en) 2013-12-05 2014-12-05 Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof

Country Status (6)

Country Link
US (1) US20170029451A1 (en)
EP (1) EP3077403A4 (en)
JP (1) JP2016540776A (en)
CN (1) CN106061982A (en)
CA (1) CA2932423A1 (en)
WO (1) WO2015085256A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108178772A (en) * 2017-10-23 2018-06-19 上海博悦生物科技有限公司 A kind of tenofovir disoproxil new intermediate and preparation method thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2685819T3 (en) 2013-11-15 2018-10-11 Chimerix, Inc. Morphic forms of hexadecyloxypropyl phosphonate esters

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101066981A (en) * 2007-05-25 2007-11-07 中国科学院上海药物研究所 Non-cyclic nucleoside phosphonate compound and its composition, prepn process and use
US20100249056A1 (en) * 2006-05-03 2010-09-30 Chimerix, Inc. Metabolically Stable Alkoxyalkyl Esters of Antiviral or Antiproliferative Phosphonates, Nucleoside Phosphonates and Nucleoside Phosphates
WO2013063436A1 (en) * 2011-10-26 2013-05-02 Chimerix, Inc. Hexadecyloxypropyl cidofovir for the treatment of double-stranded dna virus infection

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009094190A2 (en) * 2008-01-25 2009-07-30 Chimerix, Inc. Methods of treating viral infections
US20110263536A1 (en) * 2010-04-22 2011-10-27 Chimerix, Inc. Methods of Treating Orthopox Virus Infections and Associated Diseases
ES2700117T3 (en) * 2010-08-31 2019-02-14 Chimerix Inc Derivatives of phosphonate ester and methods of synthesis thereof
EP2841162A4 (en) * 2012-04-27 2016-02-24 Chimerix Inc A method of mitigating virus associated end-organ damage

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100249056A1 (en) * 2006-05-03 2010-09-30 Chimerix, Inc. Metabolically Stable Alkoxyalkyl Esters of Antiviral or Antiproliferative Phosphonates, Nucleoside Phosphonates and Nucleoside Phosphates
CN101066981A (en) * 2007-05-25 2007-11-07 中国科学院上海药物研究所 Non-cyclic nucleoside phosphonate compound and its composition, prepn process and use
WO2013063436A1 (en) * 2011-10-26 2013-05-02 Chimerix, Inc. Hexadecyloxypropyl cidofovir for the treatment of double-stranded dna virus infection

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108178772A (en) * 2017-10-23 2018-06-19 上海博悦生物科技有限公司 A kind of tenofovir disoproxil new intermediate and preparation method thereof
CN108178772B (en) * 2017-10-23 2020-01-21 上海博悦生物科技有限公司 Preparation method of tenofovir disoproxil fumarate intermediate

Also Published As

Publication number Publication date
CA2932423A1 (en) 2015-06-11
US20170029451A1 (en) 2017-02-02
WO2015085256A1 (en) 2015-06-11
JP2016540776A (en) 2016-12-28
EP3077403A1 (en) 2016-10-12
EP3077403A4 (en) 2017-08-02

Similar Documents

Publication Publication Date Title
Van Cutsem et al. Successful treatment of a squamous papilloma of the hypopharynx‐esophagus by local injections of (S)‐1‐(3‐hydroxy‐2‐phosphonylmethoxypropyl) cytosine
TWI789695B (en) Methods for treating sars cov-2 infections
CN102015613B (en) Treprostinil monohydrate
TW201819384A (en) Substituted pyrrolizine compounds and uses thereof
CN103209985B (en) Phosphate derivatives and synthetic method thereof
US10150784B2 (en) Crystal polymorph of magnesium glycinate dihydrate and process for its preparation
CN110652514A (en) Pharmaceutical use of third generation EGFR inhibitor
CN106061982A (en) Branched chain acyclic nucleoside phosphonate esters and methods of synthesis and uses thereof
US20180200257A1 (en) Methods for the treatment of inflammatory disorders
WO2019148575A1 (en) Novel use of pyridine compound
CN103330687B (en) Stable ganciclovir freeze-dried powder injection
CN113811303A (en) For the treatment of BRAFV600EThree-medicine combination of ERK1/2 inhibitor, BRAF inhibitor and EGFR inhibitor for colorectal cancer
CN1164855A (en) Antiviral nucleoside analogues containing substituted benzimidazole base attached to carbocyclic ring
CN104302651B (en) Complex compounds of germanium, methods for producing same, and drugs
US20240075038A1 (en) Application of pyrido[1,2-a]pyrimidinone analog
RU2427373C1 (en) Method for endogenous interferon induction
US20170056408A1 (en) Medicament for treating peripheral neuropathies
US20180193348A1 (en) Medicament for Treating Peripheral Neuropathies
EP3720440A1 (en) Treatment of rsv with combination product
CN108472293A (en) Conjoint therapy for cancer
CN101704795A (en) Hydrochloric acid 2-(1-ethyl propyl) moroxydine, preparation method and application thereof
CN104024247A (en) Guanidinyl-substituted polyamides useful for treating human papilloma virus
WO2022213870A1 (en) Drug and method for inhibiting cd4+treg cells by means of oral administration
US20200046715A1 (en) Medicament for Treating Peripheral Neuropathies
US10758536B2 (en) Method for treating shingles with N-methanocarbathymidine (N-MCT)

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20161026